Review

Biomarkers of aging: from molecules and surrogates to physiology and function

Published Online:https://doi.org/10.1152/physrev.00045.2024

Abstract

Many countries face an unprecedented challenge in aging demographics. This has led to an exponential growth in research on aging, which, coupled to a massive financial influx of funding in the private and public sectors, has resulted in seminal insights into the underpinnings of this biological process. However, critical validation in humans has been hampered by the limited translatability of results obtained in model organisms, additionally confined by the need for extremely time-consuming clinical studies in the ostensible absence of robust biomarkers that would allow monitoring in shorter time frames. In the future, molecular parameters might hold great promise in this regard. In contrast, biomarkers centered on function, resilience, and frailty are available at the present time, with proven predictive value for morbidity and mortality. In this review, the current knowledge of molecular and physiological aspects of human aging, potential antiaging strategies, and the basis, evidence, and potential application of physiological biomarkers in human aging are discussed.

CLINICAL HIGHLIGHTS
  • Aging is the strongest risk factor for many (chronic) diseases, frailty, morbidity, and mortality.

  • At the moment, the molecular underpinnings of aging are still only poorly understood and, accordingly, pharmacological interventions that directly target aging elusive.

  • The molecular biomarkers of aging exhibit a large variability, with very few attempts at validation in humans.

  • Physiological biomarkers of aging, centered on functional, anthropometric, and morphological aspects, are well established in large human populations, with very high predictive value for disease risks, frailty, morbidity, and mortality.

  • The physiological biomarkers, however, are somewhat underappreciated, even though they could be used in young and old healthy and clinical populations right now.

  • Lifestyle, behavioral, and environmental factors have a significant effect on human health and mortality, whereas many pharmacological and interventional approaches found in preclinical models still await human translation.

  • These factors with proven benefits should be encouraged and promoted on the individual and societal levels.

1. INTRODUCTION AND BACKGROUND

The process of biological aging has preoccupied humans throughout history, exemplified by religious lore, myths, and stories about achieving long, healthy lives, even immortality, in almost every culture worldwide, with the Mesopotamian epic of Gilgamesh as one of the earliest recorded examples, possibly dating back to 2100 BCE, in which the secret for everlasting live is desperately, and unsuccessfully, sought after (1). In comparison to prehistoric humans living as hunter-gatherers (2), human life expectancy has tremendously increased, in particular in the last 200 years, driven by various factors, including industrialization and the related abundance of food, processes that improved food preparation and preservation (e.g., those developed by Louis Pasteur), sanitation and hygiene in the public sector (e.g., drinking water treatment, sewage collection and purification) and clinical settings (e.g., hygiene promoted by Ignaz Semmelweis), as well as progress in the prevention (e.g., development of vaccines by Edward Jenner) and treatment (e.g., discovery of penicillin by Alexander Fleming) of infectious diseases and other pathologies (e.g., insulin by Frederick Banting, Charles Best, and John Macleod, general anesthesia by William Morton, and statins by Akira Endo). In addition, other factors, such as social changes including universal public health care, better education and awareness, and advances in the prevention and treatment of other diseases have also contributed to the almost unabated rise in average life expectancy, at least until recent years (3). Intriguingly, this increase was initially driven by reduced mortality at a young age, which later extended to improvements in middle and old age, thereby elevating median life expectancy, whereas maximal lifespan remained largely unaffected (46). Indeed, arguments for and against a limit of human lifespan have been put forward (4, 713), and various morphological and functional data indicate that humans might already be a long-lived species [e.g., based on resting heart rate (14), body mass (15, 16), metabolic rate (17), time of development of sexual maturity (17, 18), brain mass (17), DNA methylation rate (19) or epigenetic signature (20)], arguing for a possible lifespan maximum and a restricted window to push these limits in the absence of decisive new scientific, clinical, technological, social/societal, or ecological/environmental advances and breakthroughs (21, 22). In line with this, the current “world record” of an age of 122 yr and 164 days set by Jeanne Calment has not been broken (or even approximated) since her death in 1997 (23). In fact, at the moment, the difference between her age and that of the second oldest person, Kane Tanaka (119 yr and 107 days, death in 2022), is 3 yr and 57 days, larger than the difference between places 2 and 10 (Chiyo Miyako, 117 yr and 81 days, death in 2018) of 2 yr and 26 days.

Intriguingly, in the longest-living countries, the rise in life expectancy has been slowing down since the 1990s (24). Moreover, even though higher intrinsic capacity related to cognitive, locomotor, psychosocial, and sensory function has been measured in individuals of a certain age compared to their counterparts of earlier generations (25, 26), there is also evidence of an increasing gap between lifespan and health span (the number of years spent in good health), indicating that gains in life expectancy might not be matched with corresponding improvements in healthy aging (27). Indeed, it is indisputable that for many diseases, most of which are chronic in nature, age is by far the largest and most common risk factor (28). On a societal level, a strong demographic trend toward an aging population is observed in a number of countries (6), e.g., forecasted to triple the number of those 85 yr of age or older in the United States from 2022 to 2050 (29). This trend now extends beyond high-income countries: worldwide, the proportion of individuals aged 65 yr or higher progressed from 5% in 1950 to 9% in 2020 and is expected to continue to rise to 16% by 2050 (30). This aging segment is now the fastest growing, by 2019, for the first time in human history, having outnumbered that of children younger than 5 yr of age (31), reaching 2.1 billion individuals aged 60 yr and over by 2050 (32). Similarly, the population of 80+ yr old will expand to 426 million in the next 25 years, approximately triple that in 2020 (32). Overall, the challenges emerging from this aging trend might surpass those of overall population growth, which has declined in recent decades and is projected to continue to slow (33). Therefore, the urge to understand the aging process, and, in the optimal case, prolong human life or at least health span, is understandable in light of the functional decline, disease risk, and inevitable death faced at old age and the societal challenges that arise from the changing demographics. To do so, first, aging per se, disentangled from age-associated diseases, has to be investigated (34), since at the moment our understanding of the fundamental mechanisms driving aging is poor. For example, the concept of chronological and “biological” age as potentially diverging entities describing separate aging trajectories is still nebulous and ill-defined (3539). In fact, no consensus on the principles and processes of aging has been reached at the moment (40). Second, the definition of “health span,” or even “health” in general, should be sharpened to provide the framework for measuring and improving this important parameter (41). Good arguments exist to extend “health” and “health span” beyond the mere absence of disease and infirmity and, as suggested by the World Health Organization (WHO), include physical, mental, and social well-being (42). Such insights and advances could help to bring the “Decade of Healthy Aging,” declared for 2021–2030 by the United Nations (UN) General Assembly (43) based on an initiative of the WHO in 2020 (44), to a successful conclusion.

1.1. Aging: A Biological/Physiological Program, Stochastic Deterioration, or a Disease?

In contrast to postnatal development and puberty culminating in adulthood, generally recognized as genetically encoded and evolutionarily selected biological programs, the underpinnings of aging, in particular after reproductive age, are highly debated (45). Evolution results in the retention of favorable genetic traits in a given environment, which are only stable if passed on to the progeny. Human aging beyond reproductive age (after the menopause in women) thus could 1) be evolutionarily neutral, resulting in random deterioration and accumulation of damage; 2) be under evolutionary pressure for an accelerated process, for example to remove nonreproducing individuals from the competition for scarce resources; or 3) inversely, favor a decelerated program, allowing postreproductive individuals to care for the particularly dependent human infants, permitting adult humans to commit more time to resource provisioning by hunting and gathering (“grandmother hypothesis”) (46, 47). Survival after menopause is rare in the animal kingdom, so far described in the wild in elephants (48), toothed whales (49), and chimpanzees (50), but might be more common in mammals in captivity (51). Of note, a significant postreproductive lifespan is also observed in preindustrial humans, in absolute and/or relative length surpassing that of most animals, including nonhuman primates (48, 51, 52). Hence, unlike most species, humans substantially exceed reproductive age and exhibit remarkable longevity.

1.1.1. Insights into the aging process from long- or short-lived humans.

Unraveling the underpinnings of human aging is not easy: for example, the study of (super)centenarians is marred by the tiny sample size (53). The prevalence of centenarians is estimated at ∼1 per 2,200 individuals (of which 85% are women and 15% men) and that of supercentenarians at ∼1 per 1 million individuals (of which 90% are women and 10% men) (54). Genetic studies of long-lived individuals has led to the discovery of >50 genetic loci, albeit with small effect size (55). Most of these are linked to (cardio-metabolic) disease risk, e.g., apolipoprotein E (APOE) (55), with the potential exception of the transcription factor forkhead box O3 (FOXO3), for which the underpinnings of the impact on human aging and longevity remain to be elucidated (56). Intriguingly, centenarians, despite exhibiting a longer lifespan, have lower disease rates throughout life and thus uncouple the association of old age from the normal occurrence of major age-related diseases seen in the normal population (57). Overall, “escapers” with no clinically demonstrable disease at the age of 100 yr (∼15%), “delayers” having no age-related disease until the age of 80 yr or later (∼43%), and “survivors” who experienced pathologies before the age of 80 yr (∼42%) have been described (54). Next, so-called “Blue Zones” have been proposed as confined geographical regions with an apparent significant accumulation of healthy individuals at old age (58, 59). At the moment, no identifiable genetic signature has emerged from the study of these regions beyond potentially disease-relevant genes [analogous to those found in (super)centenarians], primarily investigated in the population of Sardinia (60, 61). Instead, the healthy longevity has mostly been attributed to lifestyle factors, including low smoking prevalence, ample physical activity, favorable nutrition, and strong social contracts (62, 63). Whereas these factors are generally applicable and accepted (see sect. 3), others are more puzzling and contrary to broader associations, for example the fact that at least some of these Blue Zones are economically disadvantaged. Moreover, in some of these regions, e.g., Okinawa in Japan or Nicoya in Costa Rica, the proposed advantages seem only valid for certain populations and are vanishing at the present time (64, 65). Whether these developments are caused by a changing environment, for example dietary habits in Okinawa (66), or if they are based on incorrect classification is currently debated (63, 67, 68). Importantly, the overall concept of such Blue Zones has been questioned because of poor recordkeeping (68, 69) and/or other causes of overinflated records of (healthy) longevity, including claims related to pension fraud (70, 71). It thus is unclear whether Blue Zones will decisively help in our understanding of aging (72). Finally, so-called “premature aging” diseases, e.g., progeria, are caused by monogenic mutations in genes of DNA repair, genomic maintenance, fidelity of DNA replication, and/or nuclear architecture, thus poorly representing the complexity and multifactorial aspects of bona fide physiological aging (73). In fact, the overall contribution of gene variants to aging is unclear: estimates for lifespan heritability range from 15% to 30% (74), whereas results from twin studies imply an even more moderate contribution (75), possibly below 10% (76). Similar to the findings in centenarians, exceptional parental longevity has been associated with reduced cardiovascular disease risk in the offspring (77). Interestingly, in such rare cases of favorable genetic endowment, benefits for health and survival are observed even with suboptimal lifestyle, socioeconomic status, or nutrition (77, 78). Heterogeneity in familial longevity, however, implies additional factors to modulate the contribution of genetic factors in heritability (79). Overall, a genetic contribution to aging, in particular to exceptional lifespans, seems highly probable, but this most likely is based on multigenetic effects with individually very small effect sizes. Nevertheless, a healthy lifestyle and other factors, discussed below, can override an unfavorable genetic endowment to a significant extent.

1.1.2. The use of model organisms to understand the aging process.

Investigating the aging process in humans is challenging because of the reasons outlined in sect. 1.1.1 and the fact that longitudinal aging studies would require decades. Consequently, human data are often based on cross-sectional associations, and molecular mechanisms of aging have primarily been studied in model organisms, with much fewer data in humans. Although lower organisms are extremely valuable for mechanistic and causality investigations, translatability of aging insights to humans might be hampered for several reasons. First, as outlined above, human aging is characterized by a long postreproductive period, and humans are potentially already reaching upper limits of longevity. Second, the most commonly used model organisms in the aging field show pivotal biological differences compared to humans (8084). For example, in Saccharomyces cerevisiae (baker’s yeast), replicative aging and chronological lifespan describe different processes for which direct human equivalents are missing. The same is true for spore formation under starvation conditions in yeast. Similarly, Caenorhabditis elegans (roundworms) can be either males or hermaphrodites, will enter a dauer stage in starvation, are prototypical postmitotic organisms in regard to the somatic cells in the adult stage (85), and initiate a self-destructive reproductive program in which somatic biomass is used at the expense of yolk production, leading to reproductive death (86). Drosophila melanogaster (fruit flies) exhibit a marked fecundity-longevity trade-off and can enter a reproductive diapause, for example when exposed to low temperatures. Starvation leads to a torpor state in Mus musculus (mouse), characterized by significantly reduced metabolic rates. In contrast, under nonstarvation conditions the metabolic rate, which is closely related to body size, is substantially higher in mice than in humans (∼7 times higher when comparing a 30-g mouse to a 70-kg human) (87). Similarly, heart rate is considerably higher in mice, with ∼600 beats/min, which is nearly 10 times that of humans (87). Furthermore, mice exhibit an ∼30–50 times faster genomic response in different inflammatory conditions (88). Moreover, mice and Rattus norvegicus (rat) have up to 10 times longer telomeres compared to humans (89). In addition, kinetics of other processes such as RNA and protein turnover are higher than the human counterparts, with protein turnover being ∼10 times faster (88). Finally, all of these model organisms have a profoundly shorter lifespan than humans, from ∼14 days (chronological) in yeast, ∼3 wk in C. elegans, ∼2–3 mo in D. melanogaster, ∼2–3 yr in M. musculus to ∼3–4 yr in R. norvegicus. As such, one human year is approximately the equivalent of 13.7 rat days (88). The most commonly used nonhuman primate model, Macaca mulatta (rhesus monkey), reaches ∼27 to maximally 40 yr and thus only up to half of the Homo sapiens (human) average lifespan. Mechanisms, interventions, and pharmacological treatments that emerge from the study of these canonical model organisms might therefore not be directly extrapolatable to humans, or not feasible because of concerns of tolerability, safety, and adverse effects and the development of tolerance, evasion, feedback mechanisms, compensation, or decompensation in the much longer timescale of application in humans. Better results might emerge from the study of other long-lived species, many of which, however, are not amenable to large-scale, controlled and standardized investigations (9093).

Besides these physiological differences, laboratory conditions might also introduce artifacts and differences compared to the environment and lifestyle of humans “in the wild.” Most canonical model organisms are bred, kept, and experimented on under strictly standardized conditions (94). For example, rodents are often housed in pathogen-reduced (or pathogen-“free”) conditions, and humidity and temperature are rigorously controlled, often not at thermoneutrality (95), below which a substantial fraction of energy intake is used to maintain body temperature (96). The circadian light-dark cycle is fixed, without seasonal variation. The animals are severely sedentary, receive ad libitum diets, and undergo health monitoring, which leads to sick animals being removed from experiment cohorts for ethical reasons, e.g., because of infections or cancer (8082, 84, 90, 97). Such environmental differences might be of uttermost importance, e.g., when trying to translate findings such as the lifespan extension in mice with interleukin 11 (IL-11) inhibition (98), which might become an issue in humans who need a fully functional immune system, in addition to the other roles of this cytokine in various tissues (99, 100). Such functions might extend to other contexts, for example the effects elicited by exercise on immune cells and function that contribute significantly to training adaptation, reduction in musculoskeletal diseases, and healthy aging (101). Environmental differences might also mask constraints of genetic effects. For example, mutations of the Methuselah gene (or antagonism of the corresponding protein) in D. melanogaster increase lifespan, but only in very specific conditions such as sex, food source, mating status, and temperature (102). Thus, the apparent longevity is not paralleled by an increase in healthy aging and is achieved at the expense of general fitness, e.g., increased susceptibility to cold, a reduction in reproductive output, and dysfunction of the neuromuscular junction (102).

Despite these caveats, model organisms have yielded insights into mechanistic aspects of aging and, as opposed to human studies, allow the acquisition of data beyond correlative or associative value. Moreover, in many regards, parallels between the physiology of model organisms and humans exist. Thus, investigations in model organisms are important and should complement human studies. Moreover, evolutionarily conserved mechanisms allude to fundamental, important molecular principles. Nevertheless, a careful validation in humans is indispensable to avoid unwarranted extrapolation. For example, generally speaking, the reduction of processes involved in growth, anabolism, and sexual reproduction and fecundity and the increase in maintenance and repair pathways have emerged as main targets to increase longevity in various model organisms (103). Thus, mice with mutations in the pituitary-growth hormone axis such as Ames or Snell Dwarf mice exhibit up to 40% increased mean and maximal lifespan, together with a delayed age-related decline in T-cell function, improved collagen cross-linking, and reduced joint cartilage degeneration as well as osteoarthrosis, better cognitive function, and lower incidence and severity of neoplasms (104). However, these benefits only manifest when mice are housed together with wild-type females since mutated males are killed by wild-type males. Moreover, cohousing is important since these animals have problems maintaining body temperature, spending a long time in energy-saving torpor. Overall, clear trade-offs between lifespan extension and physical vigor are observed (105, 106). Thus, in the wild, such mutants would be unlikely to survive and show a longevity phenotype. In this case, these findings fail to directly translate to human biology. Mutations analogous to those in Ames or Snell Dwarf mice can also occur in humans, for example in Laron syndrome. These individuals have a reduced risk for cancer and type 2 diabetes. On the other hand, they often suffer from decreased stature, prominent forehead, depressed nasal bridge, underdevelopment of mandible, truncal obesity, and micropenis in males (107). Moreover, the risk of cardiac disease mortality is increased, and more frequent deaths are reported from convulsive disorders and other non-aging-related causes (107). Most strikingly, despite a major reduction in “proaging signaling” (107), at least as defined in model organisms, mutations of the pituitary-growth hormone signaling axis in humans are not correlated with longevity (18, 108, 109). Collectively, although studies on aging processes in model organisms provide valuable insights into the molecular mechanisms, it is crucial to recognize that these organisms are not simply smaller versions of humans and validation of the findings in human cohorts remains essential.

2. PROPOSED ANTIAGING DRUGS AND INTERVENTIONS

Studies in model organisms, in some cases complemented with human data, have revealed signaling pathways, cellular processes, and key regulators to be involved in controlling longevity, so-called “hallmarks of aging.” Hallmarks of aging are defined as processes that 1) exhibit a time-dependent manifestation during aging, 2) accelerate the aging process when intensified, and 3) slow down, stop, or even reverse aging when modulated in the opposite direction (110). Twelve hallmarks of aging have been proposed: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, compromised autophagy, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis (110). Even though “hallmarks” and “pillars of aging” have been proposed, these are largely based on associative observations and fail to differentiate between causal events, epiphenomena, compensation, or decompensation in regard to human aging (111). Nevertheless, interventions and pharmacological agents have been designed and postulated to exert “antiaging” effects (112), often aiming at reestablishing dysregulated cellular properties, as defined in the aging hallmarks (FIGURE 1) (38, 110, 113128).

FIGURE 1.

FIGURE 1.The molecular and cellular hallmarks of the aging process and potential antiaging compounds. A number of molecular cellular alterations have been proposed to be associated with the aging processes, including genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, compromised autophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis (see Ref. 110). Various treatments have been suggested to target some of these processes (examples shown) and might thereby have the potential for antiaging drug effects (see Refs. 38, 110, 113128). However, at the moment, no evidence for efficacy (and safety) for the application of any of these pharmacological and interventional factors on the human aging process exists. Figure created with BioRender.com.


Examples include compounds mitigating the accumulation of reactive oxygen species (ROS)-caused damage (e.g., resveratrol, curcumin, astaxanthin, epigallocatechin-gallate, Protandim, melatonin, spermidine, and methylene blue), increased inflammation (e.g., berberine, 17α-estradiol, acetylsalicylic acid, and nordihydroguaiaretic acid), compromised autophagy (e.g., berberine, spermidine, rapamycin, and caloric restriction), impaired stem cell function (e.g., spermidine, young blood/plasma, and stem cell therapy), a decline in NAD+ levels (e.g., NAD+ precursors and boosters such as nicotinamide riboside or nicotinamide mononucleotide), excess hepatic methionine (e.g., glycine or methionine restriction), disturbed glucose homeostasis (e.g. metformin, acarbose, 17α-estradiol, and canagliflozin), cell senescence (senolytics such as fisetin, dasatinib, and quercetin, berberine, and curcumin), overactivation of mammalian target of rapamycin (mTOR) signaling, anabolism, and dysregulated proteostasis (e.g. rapamycin, spermidine, growth hormone, and caloric restriction), overactive renin-angiotensin-aldosterone (RAA) signaling (e.g., enalapril) or epigenetic drift (e.g., cellular reprogramming and rejuvenation) (38, 110, 113128). However, at the moment, none of these has been successfully tested in human aging, and the effects of most of these interventions and drugs fail to be broadly replicated even in model organisms, showing considerable species, strain, and sex differences (129). Rapamycin is one of the very few exceptions, with an effect on aging observed in different species and mouse strains (130132), with positive outcomes in the Interventions Testing Program (ITP) of the National Institute on Aging (NIA) (133, 134). Besides the conceptual conundrum of a single compound being able to alleviate multifactorial aging, other caveats exist that caution against the use of many of these drugs in humans (135). Below in this section, several examples are presented that illustrate the potential and pitfalls of “antiaging” medication. A comprehensive discussion of all proposed interventions is beyond the scope of this review, indeed might be futile considering the missing clinical data on human health and longevity at the present time. For future development, challenges for translatability of pharmacological interventions have been described (135). In addition to these, a couple of additional steps should be considered. First, reproducibility in preclinical models, as for example assessed in the ITP, might help to prioritize compounds. Second, validation in short-term randomized clinical trials should be benchmarked against biomarkers of aging with reliable data such as the physiological parameters discussed below. If positive, in terms of both efficacy of biomarker modulation as well as safety and tolerability, long-term randomized clinical trials should be done, which, even if costly, would provide data on hard clinical end points including mortality and disease risks. Moreover, such long trials would also reveal the safety, tolerability, and adverse effects profile in chronic treatment. At the moment, none of the proposed pharmacological or interventional agents fulfills these criteria.

2.1. Pharmacological Agents: Repurposed and New Drugs

2.1.1. Resveratrol.

Resveratrol is a polyphenol hailed as an activator of sirtuin 1 (SIRT1) and as a strong promoter of health and longevity (136, 137). However, initial findings could not be reproduced in mice (138), fruit flies (139), roundworms (139), or yeast (140). In fact, binding of resveratrol to SIRT1 (or the yeast ortholog Sir2) might have been an experimental artifact (140142). Moreover, the nature of sirtuins, including SIRT1, as “longevity genes” has been questioned (143). It thus was of little surprise that a multimillion endeavor of developing drugs based on the resveratrol/SIRT1 hypothesis failed (38, 143, 144). Nevertheless, resveratrol has been tested in various clinical trials for different indications, however to date with little success, controversial results, and poor evidence for efficacy (145147).

2.1.2. Metformin.

Metformin is one of the most widely prescribed medications for type 2 diabetes. This drug is in general safe and well tolerated but can, however, be associated with severe side effects including lactic acidosis, vitamin B12 deficiency, nephrotoxicity, and lower testosterone levels (148, 149). Promising initial results of lifespan extension in model organisms could not be universally reproduced (150). Similarly, early results of improved survival of type 2 diabetes patients on metformin were not substantiated (151, 152). Thus, although the usefulness of metformin in type 2 diabetes is uncontested, a potential application in healthy individuals is strongly debated (153). Intriguingly, in a 40-mo study in 12 male cynomolgus monkeys, signs of improved brain function and morphology and attenuated transcriptional fluctuations in several tissues were reported, along with mitigation of some of the proposed aging hallmarks (154). Whether these effects contribute to broad and bona fide aging benefits for health and longevity remains to be determined, as will translatability to humans (155).

2.1.3. Rapamycin.

Rapamycin, a pharmacological inhibitor of mTOR activity, was originally used as an immunosuppressant drug, e.g., in organ transplantation, but in the meantime has been applied more broadly, for example in coronary stents to prevent restenosis, lymphangioleiomyomatosis, vascular malformations, facial angiofibroma, and in different types of cancer (156). Based on the robust and highly reproducible effect of rapamycin on health span and lifespan in different model organisms, the use of rapamycin or rapalogs, compounds derived from the parent drug, in antiaging treatment of humans has been proposed (157). Surprisingly, in terms of gene expression, rapamycin treatment results in pro- and antiaging profiles, at least in skeletal muscle (158). Moreover, different muscles seem to exhibit divergent responses to rapamycin treatment (158, 159). It is now also clear that, contrary to initial hypotheses (160), rapamycin is not a “caloric restriction mimetic,” since rapamycin treatment and caloric restriction engage overlapping and distinct signaling pathways, and thus lead to at least partially divergent outcomes in the aging process (159, 161). To date, no clinical trials evaluating the effect of rapamycin on longevity or health span have been performed. However, a number of clinical trials using rapamycin or rapalogs in healthy individuals and patients suffering from age-associated diseases have led to mixed outcomes, and thus necessitate additional studies (162). Of note, most of these clinical trials were relatively short, some with a single dose of rapamycin or a rapalog. Extrapolation of tolerability, safety, and potential adverse effects to a potential longer-lasting “antiaging” treatment thus is difficult. Importantly, adverse effects occurred in these trials, even though to a lesser extent than those reported in long-term-treated kidney transplant patients (162).

Geroprotective use of rapamycin and rapalogs will thus have to be done under consideration of expected on-target and potential adverse effects. First, the inhibition of mTOR by rapamycin in B- and T-cell activation is leveraged for the immunosuppressive effect in organ transplant recipients, linked to an increased susceptibility for life-threatening infections and sepsis in these and cancer patients (163). Immunosuppression obviously would not be desired in geroprotection and might not have been seen in pathogen-shielded animal studies. However, signs of modulated adaptive immunity leading to immunostimulation in some trials indicate that the problem of immunosuppression might be preventable under certain conditions. For example, the rapalog RAD001 improved adaptive immune function, whereas the selective mTOR complex 1 (mTORC1) inhibitor RTB101 increased the risk for respiratory illness in an unsuccessful clinical trial (164). A second on-target effect is related to the antiproliferative effect on endothelial and smooth muscle cells, thereby exerting antiangiogenic outcomes, one of the main principles of rapamycin treatment of renal carcinoma (165). In fact, the deterioration of vascular function observed during aging can be ameliorated by elevating vascular endothelial growth factor (VEGF) signaling, resulting in enhanced health span and lifespan in mice (166). Hence, this effect could be negatively affected by the suppressive influence of mTORC1 inhibitors on VEGF. Other consequences of the antiangiogenic action of rapamycin include abnormal growth of the chondro-osseous junction, at least seen in rats (167), diminished wound healing (168), as well as dysregulated menses and uterine growth in women (169, 170). A more general impact on fertility is implied by gonadal dysfunction, as well as impaired spermatogenesis in men (171). Then, a broad metabolic dysregulation has been reported as unwanted response, including the development of hyperglycemia, hypercholesterolemia, hypertriglyceridemia, dyslipidemia, and diabetes (162, 172174). Finally, in preclinical mouse models, microglial mTOR has been found to enhance β-amyloid plaque clearance in an Alzheimer’s model, with inverse effects of rapamycin, indicating that pharmacological inhibition of mTOR could promote the risk for this neurodegenerative disease (175). All of these concerns will have to be addressed before long-term geroprotective treatment with rapamycin or rapalogs will be attempted.

2.1.4. Senolytics.

Senolytics target senescent cells (“zombie cells”) that are permanently proliferation arrested and characterized by the senescence-associated secretory phenotype (SASP), with potential detrimental effects on neighboring nonsenescent cells (176). Through poorly understood mechanisms, senolytics selectively remove senescent cells that accumulate in different tissues during aging, leading to functional retrieval reminiscent of younger tissue. This strategy is highly promising, with outstanding results in preclinical model organisms. However, again, human translation is still missing. Moreover, cell senescence is not merely a byproduct of aging but a physiological process that is crucial in different contexts, for example embryogenesis, wound healing, and tissue/organ regeneration (176179). Based on this knowledge and preclinical studies, a prosenescence therapy might actually be appropriate in several diseases, including pulmonary hypertension, atherosclerosis, liver and renal fibrosis, glucose intolerance, and rheumatoid arthritis, as well as cancer prevention and treatment (177). Thus, the choice of pro- or antisenescence therapy could strongly depend on comorbidities and differential diagnosis, in particular when senolytics are envisioned as geroprotective treatment in the elderly (177).

2.2. Antiaging Interventions: Back to the Roots?

2.2.1. Cellular reprogramming and (epigenetic) rejuvenation.

During embryonic development, terminally differentiated tissue cells emerge from pluripotent stem cells and originally from the omnipotent fertilized egg cell (180, 181). Since all of these cells share the same genome, the expression of the genes that define tissue identity has to be tightly regulated, most dominantly by epigenetic modifications that affect chromosomes, histones, and the DNA. Conrad Waddington’s landscape provides a theoretical framework in which stochastic alterations of epigenetic modifications and gene expression are increasingly restricted and channeled during the developmental decision-making process. This unidirectional waterfall model has been challenged and expanded. For example, dynamic changes in epigenetic modifications are observed in cell fate transitions, dedifferentiation, and transdifferentiation, e.g., in tissue regeneration (181, 182). Moreover, somatic cell nuclear transfer and the production of induced pluripotent stem cells with transcription factors provide further proof that “epigenetic barriers” can be overcome and cells reprogrammed (183). Since, arguably, reprogramming of tissue to pluripotent stem cells could be interpreted as a “rejuvenation” back to a developmentally younger version, this technique has also gained traction as a possible antiaging intervention (184, 185). In this case, potential age-associated epigenetic events, which could be either stochastic or deterministic, would be reversed. Evidence for such a drift emerges from various observations in aging tissues and organs, including loss of heterochromatin, alterations in histone posttranslational modifications and DNA methylation, accumulation of histone and chromatin modifier variants, modifications of the levels and/or activity of noncoding RNAs and transposable elements, and faster and less controlled transcription (186188). Thus, if such events were reversed, a more youthful and healthy cell function could potentially be achieved. Full reprogramming of an old organism, for example using the four “Yamanaka factors” OCT3 (also called OCT3/4 or POU5F1), SOX2, KLF4 and MYC (also called c-MYC) (OSKM), obviously would be deleterious if all cells were dedifferentiated into embryonic-like stem cells, leading to loss of organ function, severe health problems, cancer, and death within days (184). For therapeutic purposes, partial reprogramming would be the goal, to increase “stemness” without pluripotency acquisition.

Despite various successful reports in rodent animal models, human translation of reprogramming is still elusive. First, the mechanistic underpinnings of reprogramming are only poorly understood, and therefore the targeting of an exact end point in “partial” reprogramming currently is impossible. Accordingly, a large heterogeneity in outcomes is found in vitro and in vivo. In partially reprogrammed cells in culture, heterogeneity emerges from transient phenotypes/rejuvenation, epigenetic remnants and memory, loss of morphology, cell fate anomalies, or nonnatural progenitor phenotypes, the extent of which varies by method, tissue source, progenitor cell age, cell environment, and other experimental factors. A similar heterogeneity is observed in mice in vivo: abnormal tissue growth, teratoma, tumors and metastases, activation of transposable elements, hepatic and intestinal failure, and other pathological events have been reported, frequently leading to premature death (184, 189191). Again, as outlined in the discussion of drug treatment, partial reprogramming in humans must consider the much longer lifespan, which could exacerbate many of the reported adverse effects in rodents. Of note, the Yamanaka factors MYC and KLF4 can act as oncogenes, whereas the other two, OCT3/4 and SOX2, often are highly expressed in tumors (190). Inversely, several tumor suppressor genes, including p53 (encoded by TP53) and INK4A/ARF (encoded by CDKN2A) as barriers to reprogramming. Moreover, reprogramming not only is stimulated by the presence of cell senescence but also triggers this process, leading to a higher number of senescent cells and the presence of SASP in the microenvironment (184, 190). At the moment, based on our lack of deeper understanding, despite reported success in mice partial reprograming might be best described metaphorically as a blindfolded shotgun blast leading to a massive effect, but very far from clinical translation.

2.2.2. Hormones, stem cells, and young blood.

Other attempts at rejuvenation are aimed at restoring youthful function with the use of circulating hormones and other signaling factors, or with stem cells. Sex hormones such as testosterone, estrogen or progesterone, growth hormone, dehydroepiandrosterone (DHEA), and thyroid hormone have been postulated to bring back youthful features when prescribed in elderly individuals (192, 193). However, outside of clinically relevant and accepted applications, there currently is no proof for a general antiaging effect, and serious reservations about long-term effects, adverse outcomes, and risks exist.

Similarly, the injection of stem cells currently lacks evidence for antiaging properties as opposed to the well-documented use in regenerative medicine in degenerative disorders (194). Several factors might be responsible for this: for example, exogenous stem cells might not be able to fulfill their purpose if the corresponding stem cell niche is aged. Moreover, several hurdles will have to be overcome (194). Since stem cells also undergo aging-related changes that compromise function (195), autologous donor cells might first have to be “rejuvenated,” or heterologous donor cells from young compatible donors be used. The optimal type of (mesenchymal?) stem cell will have to be identified. Heterogeneity among cells and between donors, acceptors, and isolation protocols will have to be accommodated. Adequate in vitro expansion and mass production methods will have to be established to meet the very high demand for systemic treatment. Standardization of approaches, as well as methods to track the fate and function of transplanted stem cells, will have to be performed. Finally, potential long-term effects, e.g., tumorigenesis, will have to be considered.

Parabiosis experiments in rodents have shown the potential of young blood to rejuvenate organ function in connected old animals, implying the existence of circulating factors that mediate the corresponding effects (196). At the moment, the identity of such factors is not known. Moreover, based on single heterochronic blood exchange experiments, it might be possible that the extent of detrimental effects of old blood surpasses that of the benefits of young blood, suggesting that, at least in part, the rejuvenation in old animals in parabiosis might emerge from dilution of such pathological factors (197). Geroprotective benefits of therapeutic transfusion of young plasma in humans have not been documented so far. Moreover, known risks of infusion of human plasma include the presence of infectious agents, serious allergic reactions, transfusion-related acute lung injury, or overload of the circulatory system. In light of these risks, and the complete absence of proven clinical benefits for aging or most aging-related pathologies, the Food and Drug Administration (FDA) currently advises caution for the commercialization of infusion of plasma obtained from young donors (198). Moreover, ethical issues about donor recruitment and compensation and about disparities in access exist (199). Some of these might be solved once effective factors have been identified and can be recombinantly produced.

2.2.3. Caloric restriction.

A seminal study of McCay, Crowell, and Maynard (200), published in 1935, investigated whether undernutrition in rats retards growth. Intriguingly, they found that the calorically restricted and growth-retarded animals exhibited an increased lifespan. Even though this effect was only seen in males and not the already longer-lived females, this article stimulated an exponential interest in caloric restriction as longevity intervention (201). Indeed, the initial observation was replicated and expanded in various canonical and noncanonical model organisms, including yeast, C. elegans, D. melanogaster, mice, dogs, and even water fleas, silkworms, spiders, and fish (202). Because of the robustness of the effect, caloric restriction has been postulated as the “gold standard” for life-extending interventions. In a general sense, the payoff of caloric restriction on longevity decreases with animal complexity (203), from ∼200% in yeast to ∼100–200% in C. elegans, ∼100% in D. melanogaster, ∼30–50% in mice, and ∼15% in dogs [mean, not maximal lifespan (204)], with significant variations between strains and experimental protocols. To get better insights into translatability to humans, two independent trials in rhesus monkeys were undertaken, with surprisingly different outcomes (205, 206). In one study (205), an extension in lifespan was projected based on the available data, which became significant when non-aging-related mortality was excluded [estimated at ∼7% (207), thus smaller than the effects in dogs and lower organisms]. In the second study (206), no such effect was observed, and the validity of caloric restriction as longevity intervention in nonhuman primates was questioned (208, 209). Importantly, the study design differed in several key aspects (207): First, monkey breeds were not equivalent. Second, the first study had an ad libitum-fed control group, whereas the control group in the second trial was food restricted to avoid overfeeding and excessive weight gain. Third, the composition of the food was not the same, with 28.5% sucrose in the first and 3.9% sucrose in the second trial. Maybe as a direct consequence, comorbidities were unequally distributed, for example leading >40% of the control animals to develop diabetes in the first compared to 12.5% in the second trial. Thus, the positive outcome for caloric restriction on health and longevity in the first trial could primarily be based on the reduction of the pathological consequences of overfeeding of a glucose-rich diet. Indeed, in the second study, lower body mass, lower adiposity, and improved survival were already observed in the control animals (compared to the counterparts of the first study), thus even in the absence of caloric restriction, at least in the males. As a consequence, the survival of the male control cohort in the second study was the same as that of the calorically restricted males in the first and second trials, with no statistical differences. In females, no differences in body mass, adiposity, and survival were seen when comparing control and calorically restricted groups in the second study, pointing toward a sex dimorphism in the response to this intervention.

The potential benefits of caloric restriction in extending lifespan by primarily reducing pathological effects of overfeeding or unhealthy diets can also be inferred from studies in rodents. In mice and rats, the lifespan extension directly correlates with the propensity for adult weight gain, thus very little effect in lean strains and larger effects in strains that gain more weight, based either on the genetic background or on differences in food composition in the same strains (210). Similar to the outcome of the first rhesus monkey trial, many reports of lifespan extension in rodents could be due to the experimental conditions with ad libitum-overfed control mice, unhealthy dietary composition, and a marked sedentary state in normal home cages (83, 84, 97). Indeed, mice caught from the wild have lower body fat (∼3–5%) than most laboratory strains (∼9–22%, ∼22% in C57BL/6) under standard conditions (83), and caloric restriction has no effect on mean longevity in wild mice (211). Even in laboratory mouse strains, the response of lifespan to caloric restriction is far from uniform. In fact, a considerable number of mouse strains react only very little, and some even experience a negative, life-shortening effect (212214). Indeed, in genetically diverse mice, hereditability had a larger effect on lifespan compared to dietary restriction (215). Of note, in some of these mice, even in those experiencing increased longevity, dietary restriction was associated with suboptimal health, e.g., loss of lean mass, compromised immune system function, or disruption of erythroid cell populations (215). These findings are of obvious importance when considering the genetic diversity of humans (216). Moreover, as reported by McCay and colleagues in the rat strain used in their study published in 1935 (200), a sexually dimorphic response is seen in many mouse strains, which can lead to diametrically opposite effects of caloric restriction on lifespan in males and females (212214). Strain-specific responses are observed not only in rodents but also in D. melanogaster and C. elegans, in which the naturally top 10% longest-living strains obtain significantly less life-expanding benefits from caloric restriction compared to the bottom 10% (217, 218).

All of these results have high relevance when considering caloric restriction for human health span and lifespan extension, from the decreasing payoff when going from lower to higher organisms to the clear genetic and sex-specific contribution that shapes the response to this intervention. Furthermore, other issues would have to be considered, tested, and validated (210, 219, 220). For example, it is not clear what the baseline caloric intake is from which restriction is calculated. How would interindividual variations in energy metabolism be taken into account, in terms of both basal energy metabolism including non-exercise-activated thermogenesis as well as that contributed by physical activity? Then, the optimal extent of restriction, age of initiation, and dietary composition are unknown in humans. Furthermore, adherence might be compromised by the constant, unpleasant feeling of hunger (221). Finally, psychological factors of prolonged caloric restriction, e.g., on mood, depression or aggression, and reported adverse effects, e.g., frailty, reduced cognitive performance, impaired wound healing, and immune function, will have to be dealt with (94, 222). Based on the data obtained in rodents and primates indicating that the effect of caloric restriction on health span and lifespan might be rather due to an amelioration of pathologies triggered by overfeeding and sedentariness, a balanced and calorically controlled diet, linked to adequate physical activity, likely is the healthier, safer, and more efficient choice, lacking many of the potentially adverse effects, drawbacks, and limitations of caloric restriction (or other specialized diets) in humans (94). Indeed, no clear evidence of longevity benefits of bona fide caloric restriction in humans currently exists (66, 202). Moreover, similar to the nonhuman primate data, human studies such as the multicenter phase 2, randomized controlled Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy (CALERIE) trial indicate that health benefits, in this case in a young (21–50 yr) population after 24 mo of a caloric restriction of 25% from baseline calorie intake, primarily segregated to men and individuals with a higher body mass index (BMI) (223).

2.2.4. Telomere lengthening.

Other interventions have been proposed, some of which have even been commercialized even though little to no human data on benefits exist and significant adverse effects could ensue. For example, based on the shortening of telomeres in the aging process, telomerase gene therapy has been proposed as an antiaging strategy, introducing this enzyme, which can elongate telomeres, with adeno-associated viral vectors (AAVs) or cytomegalovirus vector (CMV) (224). At the moment, there is no proof of efficacy of therapeutic benefits in humans. Moreover, several caveats should put a brake on such endeavors: First, telomerase reactivation is seen in ∼85% of cancers (225). In fact, overlong telomeres are also associated with diseases in humans, e.g., familial clonal hematopoiesis (226), as well as increased cancer risk (227). The levels of telomerase after overexpression with viral vectors will be difficult to be adequately titrated in different cell types and tissues to evoke “healthy” telomere lengthening. Finally, viral vector-based gene therapy has inherent risks that might overshadow the so-far nonexistent evidence of beneficial effects in human aging (228, 229). It thus is not surprising that such “treatments,” based on telomerase, follistatin, or klotho, are not approved for clinical application in the United States or Europe for aging or related fields, and extreme caution is warranted toward commercial offers in countries with lax or nonexisting regulation. Notably, most of these suppliers do not guarantee safety or efficacy (230).

3. LIFESTYLE-BASED INTERVENTIONS IN HEALTH AND LONGEVITY

3.1. Nature: Coincidence, Genetics, and Epigenetics

In contrast to these experimental approaches, a number of interventions and lifestyle and behavioral choices have been shown to provide not only a good correlation with, but even predictive power for, human morbidity, mortality, health span, and lifespan (FIGURE 2) (231233).

FIGURE 2.

FIGURE 2.Factors and interventions affecting healthy aging, health span and lifespan, morbidity, and mortality. Nature: The genetic endowment and the benevolent outcome of random events cannot be influenced. Nurture: Lifestyle-associated behaviors can, to a large extent, be influenced on an individual level, extending to the modulation of some epigenetic modifications. Environment: Other significant factors, e.g. socioeconomic status, health care, and education, are determined by the prevailing political and societal landscape. For all of these factors, solid epidemiological and/or observational data for a significant influence on morbidity and mortality in humans exist. Yet to be identified “antiaging” drugs with validated efficacy in humans could add to or synergize with the effect of these factors. Inversely, however, they could also interfere, both on a psychological level, e.g., intake of a pill replacing exercise as an easier “substitute,” and on a mechanistic level, e.g., as described for resveratrol, metformin, or rapamycin. Thus, at the moment, efforts should center on improving the adoption and promotion of the proven factors shown here. Figure created with BioRender.com.


Apart from the fixed genetic endowment and the stochastic outcome of random events (e.g., accidents), most of the other parameters can be voluntarily changed, although some require a societal and political effort beyond the capability of an individual. Of note, genetic and lifestyle factors associate with lifespan in an independent manner, indicating that a healthy lifestyle can overcome genetic risks and convey health benefits even in a genetically “unfavorable” context (234). Moreover, environmental aspects by far exceed polygenic factors in the explanation of premature mortality (235).

The dynamic makeup of the epigenetic landscape can be inherited, predetermined, and yet be pliable. For example, the epigenetic changes elicited by the experience of famine, e.g., the Dutch hunger winter during World War II, were imprinted in the individuals who experienced this traumatic event and were transmitted to their children and grandchildren, in this case with detrimental health effects on disease risks and life expectancy (236). Obviously, you cannot chose your parents based on a benevolent genetic endowment, and you cannot influence their environmental exposures and lifestyle choices, which can result in favorable or unfavorable genetic traits and epigenetic marks for health, aging, and longevity. However, all of the lifestyle choices and behaviors discussed here will most likely result in epigenetic changes that can be beneficial or detrimental. Although for most of these factors this still has to be reliably shown, ample evidence for exercise-mediated epigenetic modifications with favorable health outcomes has been provided (237, 238). These effects can be potentiated with diet and potentially other interventions (239).

3.2. Nurture: Lifestyle and Other Factors Modifiable on the Individual Level

Importantly, the modifiable factors (including physical activity, nutrition, restorative sleep, no excessive alcohol, no smoking or opioid use, stress management, and social connections) are not mutually exclusive, and additive or synergistic effects can be achieved when they are combined (240253), even at old age (254) or in regard to various diseases such as dementia (255, 256), frailty (257), diabetes (258), brain health (259, 260), stroke (261), and cancer (262). For example, whereas adopting one lifestyle factor reduces the mortality risk by 26%, the implementation of eight factors results in a lowering of the mortality risk of 87% (241). Of these, physical activity is by far the best intervention [“Exercise is Medicine” (263)] and is modifiable on a personal level with clear benefits for health and longevity (264284), far beyond cardiovascular mortality (285289) and regardless of age (290) or sex (291, 292). Health benefits of physical activity were already proposed in ancient cultures, two and more millennia ago (293, 294). Exercise, like aging, is multifactorial, affecting almost every organ and tissue in the human body (295). It thus is not surprising that all of the proposed hallmarks of aging are ameliorated by this intervention (296299). The underpinnings of this potent effect are, however, unclear: one hypothesis proposes that the stress elicited by an acute exercise bout mimics some of the changes observed in aging and, if done repeatedly in training, provokes a better protection and resilience against these processes (300). However, it is noteworthy that despite an apparent risk-protection paradox, for example due to the marked stress exerted on the cardiovascular system during an endurance exercise bout, regular physical activity reduces mortality risks and optimally prepares the body for ensuing perturbations in an effective and long-term sustainable manner (301). Exercise accordingly rivals existing drugs in terms of efficacy to prevent and treat a number of pathologies (302304). Inversely, a sedentary lifestyle is a strong and independent risk factor for many chronic diseases and mortality (264, 305309). Of note, most data on the effect of physical activity on mortality are derived from observational studies as opposed to the very rare randomized clinical trials with mortality as specified primary end point (310). Nevertheless, the wealth of data indicates a higher probability for a direct, causal relationship compared to mere association (310). Physical activity and exercise are discussed in more detail in the sections below.

Dietary patterns and nutrition scores are likewise associated with frailty and mortality (311, 312). However, the effect of different diet modalities and macronutrient enrichment in human aging remains debated (94). Moreover, rodent studies, e.g., showing a beneficial effect of low-protein and -amino acid diets on longevity, might not be extrapolatable to humans (94), in whom anabolic resistance necessitates higher protein supplementation to mitigate age-associated muscle mass loss and frailty (313318) and in whom high-protein diets might improve sarcopenia (319) or mortality, even in patients with chronic kidney disease (320). Such effects of higher protein intake are significantly boosted by concomitant resistance exercise training (321). In any case, it is clear that a balanced [for all necessary macro- and micronutrients (322, 323)] and calorically controlled diet, possibly devoid of ultraprocessed food (324), is crucial to lower the risk for obesity and cardiovascular and metabolic disorders, thereby helping to maintain health from young to old age (325327), in combination with adequate hydration (328). The higher relevance of overall healthy eating patterns over specific macronutrient depletion/enrichment was accordingly highlighted in the Dietary Guidelines for Americans (DGA) 2020–2025 (326). Moreover, the impact of healthy food choices on mortality has been demonstrated (329, 330), in particular in but not limited to elderly individuals, who are often plagued by malnutrition and inadequate protein intake (331). In the future, individual differences in food absorption, metabolism, and excretion might be leveraged to design precision/personalized nutrition, conferring additional health benefits (332, 333). However, better ways of monitoring and recording dietary habits will have to be devised to circumvent often unreliable self-reporting (334, 335).

The optimal amount of sleep is age dependent, with a recommended duration of 7–9 h per night for young adults and of 7–8 h per night in older adults, following a healthy pattern with few interruptions, insomnia, snoring, or other events that lower sleep quality (336, 337). Both shorter (<6 h) as well as longer (>9 h) sleep are associated with an increased mortality risk, most prominently for cardiovascular but also other types of mortality (338343) or dementia (344). Similar to most other lifestyle interventions, sleep is interdependent on other lifestyle and societal factors, for example socioeconomic status, depression and further psychiatric issues, alcohol dependence, and sedentary behavior (345). Interestingly, mortality associated with overlong sleeping seems to be predominantly affected by such environmental factors, whereas that of shortened sleeping exhibits a stronger heritability component (346). Overall, sleeping patterns not only contribute to the prediction of body characteristics (e.g., visceral adipose tissue) and disease risks (e.g., insulin resistance) but can inversely also be inferred by lifestyle factors to >50% (347). Importantly, changes in sleeping behavior toward optimal patterns can reduce mortality risks (338), even when performed in the form of weekend catch-up sleep (348). Sleep quality can for example be promoted with endurance or resistance training (349, 350), in normal sleep as well as in the context of sleep disorders (351). At least somewhat related to sleep patterns, disruption of circadian rhythms with bright nighttime and dimmed daytime light is sufficient to increase mortality risks (352).

In the elderly, poor cognitive performance is an independent risk factor for mortality (353, 354). Unfavorable results in cognitive tests could be the consequence of an unhealthy lifestyle or comorbidities, with limited effects of cognitive training in old age (355). A causal relationship between sarcopenia and cognitive impairment (356) and a correlation between physical activity levels, muscle strength, working memory, and cognitive function have been postulated (357). Nevertheless, despite studies showing positive effects in healthy individuals (358), the efficacy of exercise to improve cognitive abilities in the elderly, in particular with already compromised function, is controversial (359364) but might be boosted by multidomain interventions that include physical activity (365, 366). When undertaken at earlier age, interventions, e.g., focused on healthy diet, physical activity, and abstinence from smoking, might help to build a cognitive reserve, with clear benefits of lifetime intellectual and cognitive engagement (355, 367, 368). Of note, skeletal muscle mitochondrial oxidative capacity, a parameter that is highly pliable by endurance training, is associated with preserved brain structure, in particular of areas involved in cognition, motor function, and sensorimotor integration, hinting at a correlation of exercise adaptations and muscle mass with brain health and structure (369376). Finally, early detection and monitoring of trajectories could be improved by combining functional (377), imaging- and blood biomarker-based approaches (378) and might help to distinguish interindividual differences present from birth/young age from those arising during aging (379).

Preventative health checks and monitoring can contribute to the risk assessment for various diseases and disease-specific and overall mortality (380), e.g., by periodical recording of blood pressure, body mass as well as waist-to-hip ratio, and cholesterol, lipid, and glucose levels. However, for many screenings clear benefits have not been established, and problems with false positive and false negative findings can arise. Thus, in a healthy population, random general health checks might not help to decrease mortality risks (381). In contrast, well-designed, evidence-based health monitoring, combined with an up-to-date vaccination status (382, 383), will help in the prevention, early detection, and treatment of pathologies, thereby reducing mortality risks (384, 385). Moreover, such programs can trigger healthy lifestyle behavioral changes in the monitored population (386).

Risk behavior can be a strong driver of mortality, for example in adolescents (387). However, also in later stages throughout life, engagement in risky behavior such as violence, substance abuse, unsafe sexual habits, or reckless driving highly increases the chance of death (388). In a number of countries including the United States, gun ownership is likewise associated with a higher mortality risk, with an influence on both homicide as well as suicide rates (389392) and constituting one of the leading causes of death in children and adolescents (393). This is one of the reasons why life expectancy in the United States lags behind that of other rich countries (394).

Smoking still is one of the strongest modifiable drivers of premature mortality, even though smoking rates are declining in many countries (395, 396). Interestingly, smoking habits could contribute to the sex differences in life expectancy (397, 398). The abuse of other recreational drugs likewise increases mortality risks. Alcohol, for example, not only can lead directly to fatal pathological events (399) but also increases risk behavior such as reckless driving, other violent and nonviolent injuries, and accidents and can exacerbate psychiatric disorders and suicidal behavior (400, 401). Similarly, the opioid crisis in the United States leads to an estimated 3.1 million years of life lost (38 yr per death) and contributed to the decrease in life expectancy between 2019 and 2022 (389, 402).

With regard to social and mental factors, psychosocial stress and stress-related disorders are associated with the risk for several chronic diseases and increased mortality (403406). Intriguingly, a bidirectional association between sedentary behavior and psychosocial stress has been reported (407). In contrast, optimism shows a positive association with cardiovascular events, mortality, and longevity (408410). The amount and quality of social interactions are also predictors of mortality (411, 412). Loneliness and isolation promote stress, whereas interpersonal helping behavior decreases stress-related mortality risks (413415). As with other lifestyle factors such as overlong sleep, loneliness could constitute a causal or a surrogate marker, being potentially indicative of comorbidities such as frailty (416), anorexia or sarcopenia (417), socioeconomic constraints, or other confounders (418).

All of these lifestyle factors can be influenced by behavioral decisions. To a certain extent, this is also the case for sun exposure, although geographical and climate-based limitations exist. Insufficient sun exposure increases the risk for many pathologies and ultimately leads to a significant number of preventable deaths (419421). Notably, many of the health benefits of adequate sun exposure are independent of vitamin D production, and supplementation falls short in preventing the pathological outcomes of insufficient sun exposure (419, 420). Obviously, the harms of excessive sun exposure, in particular the development of skin cancer, should be avoided, such as sunburns or inadequate protection of eyes and skin at times or seasons with high ultraviolet radiation levels (419, 420, 422, 423).

3.3. Nurture: Societal and Political Aspects

The opportunities for a change in socioeconomic status depend on a variety of factors, many of which, for example access to high-quality and affordable education, fair income and taxation systems, or absence of discrimination and “glass ceilings,” can only be achieved on the societal and political levels. Educational disparities (424) and low socioeconomic status increase mortality risks even when adjusted for other risk factors (425428), with up to 20 yr of life expectancy disparities between poor and wealthy areas in the United States (429, 430). In fact, at least in the United States, of all social determinants, socioeconomic factors have been proposed to contribute most to health outcomes and length and quality of life (∼40–47%), surpassing health behaviors (30–34%), clinical care (16–20%), and the physical environment (3–10%) in importance (431, 432). It is noteworthy that these effects apply to years lost and years of functioning lost (433). Moreover, accumulation of the consequences of low socioeconomic status on health and life expectance over the course of a lifetime has been proposed, with consequences of experience in childhood affecting adult health (434). Thus, the childhood postcode can, at least in certain countries like the United States, predict lifetime risks for many pathologies reasonably well (435), together with other environmental factors, much better than contemporary polygenic scores (235, 436). Strategies to overcome socioeconomic hurdles and provide the means to achieve a healthy lifestyle have been proposed but, in many cases, remain to be implemented (437, 438).

Socioeconomic status might also limit the availability of or access to affordable high-quality health care in countries in which this is restricted, e.g., those without universal health care (233), or where inequalities in access such as to primary care physicians exist (439) or differences in health care spending are observed (440). Such disparities lead to a high number of preventable deaths (441, 442). Most prominently, vast differences in global probability of premature death are observed, in part driven by infectious and maternal health conditions more prevalent in certain regions, e.g., sub-Saharan Africa, than others (233, 443). Among the Organisation for Economic Co-operation and Development (OECD) high-income countries, the United States is an outlier in terms of health care costs and outcomes (444449). When expressed both as per person and as percentage of gross domestic product (GDP), health care spending is substantially higher in the United States compared to the other countries. Even though the situation has improved with the historic passing of the Affordable Care Act, a significant proportion of the population in the United States remains uninsured, whereas in the other OECD countries health care is mandatory through public and/or private programs. Despite the higher spending, life expectancy at birth is 3 yr lower in the United States compared to the OECD average, and the share of avoidable deaths (normalized to 100,000 people) is higher, including the highest OECD rates for infant and maternal mortality (450, 451). These data illustrate that even in countries where the highest-quality and cutting-edge medical care is available for those who can afford it, socioeconomic barriers and ethnic/racial disparities exist, with significant consequences on overall health and mortality outcomes (452, 453). Finally, as recent events have shown, health care policy should include an agenda for pandemic prevention, preparedness, response, and recovery/reconstruction on the national as well as global level (233). In fact, a chance of >20% for a pandemic to occur in the next 10 years that will kill as many individuals as COVID-19 has been estimated (233). Related, but not limited to the COVID-19 pandemic, high-quality education should not only aim at improving knowledge on the basis and application of all aspects of a healthy lifestyle but also convey methods for critical thinking, assessment, and classification of scientific evidence and hypotheses, facts, mis- and disinformation, and fake news (454, 455).

Changing the consequences of anthropogenic climate change and air pollution is beyond the capabilities of individuals and will require a coordinated effort on national and global scales. Nevertheless, clear effects on mortality have been reported. For example, the increasing fluctuations and extreme temperatures caused by human-made climate change will lead to deaths beyond affecting only the most susceptible populations, those with low socioeconomic status, infants, and the elderly (456459). Even in moderate climate zones, particulate air pollution is strongly associated with all-cause, respiratory, cancer, cardiovascular, and other types of disease-specific mortalities (460, 461), as well as with brain health (462). This can be synergistically exacerbated by high temperatures (463). Other types of pollution, for example environmental microplastics (464468), endocrine-disrupting chemicals (469, 470), and (transportation) noise (471473), might exert similar effects on disease and mortality risks.

4. AGE-REVERSAL-AGE-EXTENSION PARADOX: MORE MIGHT NOT BE BETTER

4.1. Interference between “Antiaging” Drugs and Epigenetic Programming

For the lifestyle- and behavior-based factors that influence mortality, additive or synergistic effects have been shown, in the absence of adverse interactions or side effects. This seems to be somewhat different for the proposed preclinical treatments and interventions, in which some combinations result in additive or synergistic outcomes (474) whereas others could be irreconcilable. For example, the age-reversal-age-extension (ARAE) paradox has been proposed for epigenetic programming, in which drugs that promote genomic stability are incompatible with the reprogramming that flattens the epigenetic landscape to enable upward movement in the Waddington landscape (184). Such effects have been shown for some of the proposed “antiaging” drugs that interfere with epigenetic reprogramming, including metformin starting at a concentration of 10 µM (age interventions are often tested at 100 µM), rapamycin starting at 1–2 nM (human plasma concentrations in rapamycin-treated patients range from 5 to 30 nM, and 50 nM is often used in longevity studies), and resveratrol starting at 20 µM (most health benefits in model organisms are seen with concentrations of 10–25 µM) (184). Thus, even if clinical benefits were achieved, potential interactions might preclude combination-based approaches of these proposed therapies.

4.2. Many of the Proposed “Antiaging” Drugs Dampen Exercise Training Adaptation

Additionally, the ARAE paradox or analogous paradigms can be expanded to other unfavorable interactions. As described above, epigenetic programming is inversely associated with cell senescence, thus facilitated by the presence of senescent cells and itself triggering cell senescence (184). The outcome of a combination therapy based on reprogramming and senolytics therefore is uncertain. These issues are theoretical since all of these approaches are still experimental. More concerning are interactions with proven lifestyle interventions that have been reported in human trials. These have been mostly shown for exercise (37, 38, 475, 476) but might also apply to others. For example, resveratrol blunts the positive effect of endurance exercise on cardiovascular health and reduces the training effect by ∼45% (477479). Metformin dampens adaptations to endurance (480, 481) and resistance (482, 483) training by ∼50% and increases the rate of perceived exertion (484). Antioxidants (485), e.g., high doses of vitamin C and E, reduce the positive effect of exercise on insulin sensitivity (486), delay recovery from endurance (487), and diminish peak torque and total work in resistance training (488, 489). At least in mice, nicotinamide mononucleotide (NMN) reduces exercise benefits on hepatic triglyceride accumulation, insulin secretion from islets, and glucose tolerance in diet-induced obesity (490). Moreover, NMN might be associated with other adverse effects, in particular in certain patient populations (491, 492) or the elderly (493). Finally, inhibition of mTORC1, a key regulator of anabolism in resistance training (494496), with rapamycin reduces stimulation of skeletal muscle fractional protein synthesis rates and growth in experimental settings of muscle hypertrophy in rodents (497, 498) and in resistance exercise in humans (499). Interference with endurance training has not been tested so far. However, mTOR is also activated in this type of training modality (500). Moreover, one of the key therapeutic targets in rapamycin therapy in cancer, a reduction of angiogenesis and neovascularization (165, 167), could be relevant for endurance exercise-induced vascularization of muscle tissue, an important process in endurance training adaptation (295). Indeed, adverse outcomes in other types of physiological angiogenesis have been reported, e.g., uterine growth and menses or wound healing, in which this process can be impaired in rapamycin-treated patients (168170). Mechanistically, at least in part, the antiangiogenic effect of rapamycin has been attributed to a positive effect of mTORC1 on hypoxia-induced factor 1α (HIF-1α) (165). Since different exercise modalities result in reduced physoxia in skeletal muscle tissue, HIF-1α plays an important role in exercise adaptation (295), which could be impaired by rapamycin administration. Such potential harmful interference of drugs with exercise obviously should be avoided (37, 38, 97, 475, 476, 501, 502). For some, adjusted dosage and timing have been proposed to minimize potential antagonistic effects on exercise adaptation. For example, it has been speculated that restricted use of rapamycin on nontraining days could mitigate adverse effects on muscle protein synthesis stimulated by resistance exercise. This however is questionable, and needs to be critically tested, in light of the long anabolic window with increased muscle protein synthesis (up to 48 h and more) after a resistance exercise training bout (503, 504). Other factors should furthermore be considered. First, it is unlikely that the benefits of single compounds exceed those elicited by the multifactorial adaptations in exercise. Thus, failure to achieve optimal health outcomes might ensue because of the mitigation of the broad and proven effects of exercise that is not outweighed by the narrow effects of single pharmacological compounds. Second, even besides potential direct interactions, the use of an “antiaging” drug could lead to reduced adherence to and compliance with lifestyle interventions, including physical activity or a balanced diet. Psychological effects of a false sense of “pharmacological health,” seemingly induced by geroprotective agents and putative “exercise” or “caloric restriction mimetics,” might diminish the motivation to engage in time-consuming, long-term, and arduous activities if an attractive alternative seems as easy as taking a daily pill.

5. MOLECULAR BIOMARKERS OF AGING

Many of the problems that affect the study and potential treatment of human aging boil down to the long time frame that is needed based on human life expectancy, as well as on our ignorance of the evolutionary and molecular underpinnings of this process (505, 506). Following large cohorts over years to decades in prospective clinical trials necessitates a high financial and logistical commitment. However, aging, a physiological (?) process experienced by every human, is not recognized as a disease by regulatory agencies, at least not at the moment. Accordingly, treatment and trials would by definition be performed in “healthy” individuals, with very high regulatory and ethical standards. To get around these issues, model organisms with shorter lifespans and lower ethical hurdles for experimentation are studied (80, 81). Furthermore, human aging biomarkers as surrogates to measure more short-term effects of treatments on aging progression are being investigated (506510).

5.1. Molecular Clocks: from Epigenetics to Metabolites

A number of such molecular biomarkers of aging have been proposed, from epigenetic marks to telomere length, transcriptomic and proteomic signatures, and glycoprotein profiles (506, 508, 511515) (FIGURE 3).

FIGURE 3.

FIGURE 3.Molecular biomarkers and aging clocks. The age dependence of several molecular events is leveraged to predict whether “biological age” deviates from chronological age. Processes that are monitored range from telomere length to epigenetic marks, most notably DNA methylation events. Specific fingerprints of transcripts, proteins, metabolites, and protein glycosylations and glycation have likewise been proposed as biomarkers for aging. Most of these age-dependent molecular changes occur in all cells, tissues, and organs of the body. However, significant differences exist between individual tissues/organs, between cell types, or even between cells of the same type based on spatial organization within an organ. One way to avoid potential confounding effects of cell heterogeneity is to define plasma metabolite or plasma protein profiles that are associated with aging. Of note, some of these clocks are affected by circadian rhythms or external perturbations such as pregnancy, infection, diseases, and potentially other factors such as chronic stress. AGE, advanced glycation end product. Figure created with BioRender.com.


Epigenetic clocks are based on the observation of changes in epigenetic modifications from embryonic development to aging. As described above, epigenetic programming attempts to roll these back to a more youthful state. Initially, DNA methylation events measured in saliva or blood samples were used to establish models to predict “biological age.” Later, epigenetic clocks were also measured in other tissues and cell types. At the moment, a lack of understanding of the molecular causes and consequences of these DNA methylation changes hampers obtaining mechanistic insights beyond the current correlative value. Recently, histone marks of aging have been proposed to perform with predictive power similar to epigenetic modifications (516). Transcriptomic clocks rely on the assessment of age-dependent gene expression profiles, e.g., in peripheral blood mononuclear cells or dermal fibroblasts. Most of these clocks, however, have only been tested in small cohorts and are marred by inherently large transcriptional noise, in particular in aging.

Advances in mass spectrometry and antibody- or aptamer-based techniques have facilitated the development of proteomic clocks. Plasma protein signature, however, can be affected by organ function, e.g., that of the kidney. Nevertheless, recent attempts at establishing blood plasma protein biomarkers based on large human cohorts have shown promising results in terms of predictive power for organ health, morbidity, and mortality (517529). Indeed, of all the molecular/-omics clocks, plasma proteome biomarkers seem most advanced, both technologically as well as in terms of high-throughput human application and validation (530, 531). The continuous improvements in detection sensitivity and proteome coverage of mass spectrometry and DNA aptamer-/antibody-based platforms, combined with machine learning algorithms, will help to eventually push this approach to broad clinical application, even though costs currently are still prohibitive (530). Metabolomic clocks (532) have likewise benefited from better mass spectrometry methods, as well as state-of-the-art nuclear magnetic resonance (NMR). However, many of the metabolites identified by NMR are of unknown structure, limiting the usability of untargeted metabolomics, further exacerbated by the noise in measurement. Nevertheless, metabolomics studies that include physiological biomarkers show promising associations in longitudinal human trials (533). Compared to these approaches, attempts at creating clocks based on protein glycosylation, advanced glycation end products (AGE), chromatin marks, or state have so far been more limited. Finally, as an example of a non-omics-based clock, telomere analysis determines telomere length as a function of age (534). Methodological issues, however, can lead to considerable variation, between both individuals and tissue types or even between sampling sites in the same tissue (or heterogeneity between peripheral blood mononuclear cells) (535).

5.2. Molecular Clocks: Current State and Challenges

At the moment, no consensus on methodology and clock as standard biomarker for human aging has emerged. For most of these biomarkers, the physiological and functional relevance are unclear, the predictive value for morbidity and mortality in humans is poor, and there is little overlap between different types of clocks, and even between clocks of the same type (507, 508, 536, 537). For example, in a recent study investigating the effect of long-term caloric restriction in healthy adults on DNA methylation, the DunedinPACE clock showed a slight reduction in “biological age” in the restricted group, whereas two other clocks (PhenAge and GrimAge) did not produce this effect (538). Significant deviations between technical replicates of the same samples can exist, in one study with median and maximal deviation of 3 yr and 8 yr, respectively (539). Another study of six epigenetic clocks using the same samples resulted in deviations of up to 9 yr (540). Moreover, epigenetic clocks differ between organs and cell types (541, 542), even between the same cell type depending on spatial location within a tissue (543). In fact, some tissues, for example skeletal muscle, are only poorly represented by epigenetic clocks (544). Then, time of sampling can affect the results, as many epigenetic clocks exhibit circadian oscillations (545). Furthermore, results obtained in model organisms might be misleading. For example, the naked mole-rat is considered a demographically nonaging animal, with exceptional longevity and almost complete absence of cancer and cardiovascular diseases. Nevertheless, this animal exhibits epigenetic aging (546). Such findings might indicate that many of the epigenetic events that are measured might not correlate with bona fide aging but rather represent physiological and pathological consequences of different types of events. In fact, epigenetic clocks often result in overlapping outcomes when comparing old and cancer cells (508). Moreover, the transient modulation and subsequent regression of DNA methylation marks in pregnancy or infection might not be a sign of increasing and decreasing “biological age” (547) but a normal response to these types of stressors (548). Hence, it is essential that a biomarker is robust and not affected by acute physiological perturbations or by technical or preanalytical variability. Thus, at the moment, all of these molecular clocks will have to be used with caution, awaiting validation in longitudinal human studies in large cohorts (549, 550).

Efforts to standardize the development and validation of such clocks are currently underway and will be indispensable for breakthroughs in this area (506, 551). More specifically, different types of biomarker validation have been proposed that might help to improve the discovery and implementation of biomarkers (BOX 1) (506, 551).

BOX 1. Proposed framework and criteria of aging biomarkers (adapted from Ref. 506)

Classification: How can the biomarker be used?

1) 

Predictive (e.g., for aging trajectories, identification of individuals who respond to interventions)

2) 

Prognostic (e.g., trajectories and treatment of age-associated diseases)

3) 

Response (e.g., biological reaction to aging or interventions)

4) 

Surrogate end point (e.g., to substitute for direct measurement of aging)

Assessment: How can the biomarker be measured?

1) 

Feasibility and validity of measurements (e.g., prerequisites for sample acquisition, processing and data interpretation, age sensitivity, non-age-accelerating)

2) 

Mechanistic underpinnings and biological plausibility (e.g., reflection of underlying mechanisms involved in the aging process)

3) 

Generalizability (e.g., narrow vs. broad applicability from model organisms to humans, usage in different ethnicities, sexes, age groups, and other demographics and populations)

4) 

Response to aging and interventions (e.g., reflecting improvement or deterioration in the aging process and, inversely, promotion of geroprotective effects)

5) 

Costs (e.g., application in large-scale settings and lifelong longitudinal assessments)

6) 

Invasiveness and safety (e.g., minimally invasive, devoid of adverse effects, safe to test from the young to the oldest of the old)

Validation: Has the biomarker been validated in terms of measurement methods and clinical application?

1) 

Analytical validation (e.g., quantifiable, accurate, reliable, repeatable, and reproducible measurement, standardized procedures)

2) 

Clinical validation (e.g., validity in different human cohorts, statistically relevant reference values for broad demographics and populations)

First, biomarkers are especially valuable when they originate from pathways that actively drive aging (biological validation) rather than simply being correlated with this process. Second, if a biomarker targets conserved pathways and is validated in multiple species (cross-species validation), it is more likely to be linked to the fundamental process of aging. Third, the predictive value of a biomarker for future age-related outcomes (predictive validation) in a prospective rather than retrospective study should be assessed. Fourth, the assessment/analysis of the biomarker should be standardized (analytical validation), including the collection, storage, and analysis procedure of the sample to facilitate the determination of the sensitivity, specificity, and reproducibility of the biomarker. Finally, it is important to establish the clinical value (clinical validation) of a biomarker in terms of improving our understanding of the disease or the potential effects of an intervention on health outcomes. Collectively, broad validation across these dimensions is instrumental to ensure that molecular biomarkers are robust, reproducible, and clinically meaningful and can therefore be used to predict “biological age,” health outcomes in response to an intervention, and the risk to develop diseases and of mortality.

Furthermore, multi-omics approaches might help to improve such clocks (552), analogous to attempts in the field of muscle exercise biology (553). However, even with the current lack of a comprehensive understanding of the aging process, a multicomponent mechanism, with pleiotropic outcomes, affecting all cells, tissues, and organs of the human body, is more likely than a single pathway/molecule common to all somatic cells. Accordingly, until our knowledge improves, compound measures and indices that describe systemic, or at least multiorgan, health, relevant for resilience and functional retention, might have the highest chance to monitor aging progression and provide feedback on potential interventions (discussed below). Nevertheless, it is conceivable that the study of these molecular clocks will help us to better understand the mechanisms underpinning human aging. Moreover, the use of integrated or combination approaches, e.g., combining plasma proteome with human phenome data (554) or metabolome with genomic information (555), and the increasing use of machine learning of large datasets might help to improve the quality of these clocks (508, 530, 552, 556558). Attempts at integrating physical fitness or other lifestyle-/behavior-related parameters in epigenetic clocks (559561), however, might be better served by measuring the real thing instead of a proxy of uncertain value. In fact, clinical “aging clocks,” based on anthropometric, (patho)physiological, and molecular parameters, have been proposed to predict healthy and unhealthy aging trajectories (562, 563). In any case, proven, physiological biomarkers of aging should be used to benchmark any new molecular biomarkers (564567).

6. PHYSIOLOGICAL BIOMARKERS OF AGING: READY FOR PRIME TIME!

As outlined above, physical activity remains one of the best interventions for human health and longevity. It thus is not surprising that the assessment of morphological, anthropometric, and functional parameters that describe the outcome of exercise training provides strong predictive power for health, morbidity, and mortality. In fact, these physiological biomarkers of aging provide an assessment of functional aspects (568), relevant in everyday life, e.g., on fatigability, strength, or gait speed, that affect daily tasks such as walking across a pedestrian crossing, moving up stairs, carrying groceries, having social interactions, or being able to clean the apartment, thereby markedly impacting on quality of life and independence (569). Despite proven associations (506) and current indispensability as benchmarks in preclinical as well as end points in clinical trials (570), these parameters surprisingly often are underappreciated in contemporary discussions of biomarkers of aging. Thus, to increase the recognition of these measures, and to draw a direct comparison, the physiological biomarkers are positioned in the proposed framework of aging biomarkers (BOX 1, adapted from Ref. 506) in this review. As outlined below, this will show that the physiological biomarkers of aging fulfill the requirements and challenges that have been put forward (551). Broadly, the physiological biomarkers of aging can be categorized in terms of cardiorespiratory fitness, muscle mass, muscle strength and power, leisure-time activity, neuromuscular function, and frailty. The most common parameters and tests are described in more detail below in this section. Of note, the deterioration of these tissues and organs, combined with neurodegeneration and a reduction in bone mass and mineral density, is universally observed and thus could be considered true “physiological hallmarks of aging,” with a clear and causal relationship to the decline in functional capacity, morbidity, and mortality (FIGURE 4).

FIGURE 4.

FIGURE 4.Physiological/functional hallmarks of aging. The loss in muscle mass, often associated with a change in fat mass and distribution, reduced cardiorespiratory fitness, impaired muscle strength/power, and neuromuscular deficiencies associated with frailty, are age-related processes that are observed universally, all of which are measured with the physiological biomarkers of aging. In addition, reduced brain health, in particular driven by neurodegeneration, and a loss in bone mass and mineral density occur. Most of these hallmarks of aging can be determined by the proposed physiological biomarkers. Of note, all of these hallmarks of aging are ameliorated by lifestyle- and behavior-based interventions, most importantly exercise training. CT, computed tomography; DEXA, dual-energy X-ray absorptiometry; MRI, magnetic resonance imaging; V̇o2max, maximal rate of oxygen consumption. Figure created with BioRender.com.


6.1. V̇o2max/Cardiorespiratory Fitness

6.1.1. V̇o2max: principle and testing.

At the moment, cardiorespiratory fitness is the most studied and best predictor of morbidity and all-cause as well as disease-related mortality (FIGURE 5, A AND B) (572583). Cardiorespiratory fitness is assessed by measuring the maximal rate of oxygen consumption (V̇o2max). This concept was introduced by Archibald Hill and Hartley Lupton in 1923 (584) and has been developed ever since (585, 586). V̇o2max, sometimes also reported as V̇o2peak [the highest recorded V̇o2 in tests failing to reach a V̇o2max plateau, therefore potentially underestimating the true maximum (587, 588)], is a readout obtained in cardiopulmonary exercise tests that correlates with cardiorespiratory fitness and endurance capacity (589593). Most often, V̇o2max, either reported as an absolute rate (mL/min) or normalized to body mass (mL/min/kg) (sometimes also normalized to lean body or skeletal muscle mass), is typically determined in graded maximal exercise tests by measuring ventilation and respiratory O2 concentrations (reaching a plateau), often combined with determination of blood lactate concentrations (e.g., approaching or exceeding 10.0 mmol/L), heart rate (reaching a plateau), respiratory exchange ratio (RER; ≥1.1), and perceived exertion [e.g., 19–20 on the Borg scale from 6 to 20 or 9–10 on a Borg Category-Ratio 10 (CR10) scale from 1 to 10], even though variations between protocols for some of these thresholds exist (594596). The results depend on the exercise modality: treadmills or cycle ergometers are commonly used, but V̇o2max values can be acquired in any exercise setting that is amenable to breath-based oxygen analysis, e.g., rowing ergometers, all of which involve different sets of muscles. The V̇o2max often differs between testing modalities and prior task habituation: for example, runners might reach higher values on the treadmill, whereas cyclists or triathletes can excel on cycle ergometers (597). The choice of testing paradigm therefore depends on availability, practicality, intention for assessing task specificity, coadministration of other tests (for example, electrocardiography might be easier on cycle ergometers because of minimal upper body movement), and other factors.

FIGURE 5.

FIGURE 5.Maximal rate of oxygen consumption (V̇o2max) is a strong predictor of health, morbidity, and mortality. A: V̇o2max represents a multisystem readout of cardiorespiratory fitness, which is highly pliable by exercise. The age-associated decrease of up to 10% per decade can meet an aerobic frailty threshold, leading to disability and loss of independence. V̇o2max is substantially higher in elite athletes, with measures ∼2-fold above those of the general population. B: age-related V̇o2max in a cross-sectional patient study of different ages and sexes (n = 122,007; men: n = 72,173; women: n = 49,834), acquired over 24 yr, tested by treadmill running. The 25th, 50th, and 75th percentiles for each sex are indicated by solid (50th) and dashed (25th and 75th) lines. Adjusted all-cause mortality hazard risk ratios between groups (pooled over all age groups and both sexes) are all significant (P < 0.001). Groups: low (<25th), below average (25th–49th), above average (50th–74th), high (75th–97.6th), exceptional (>97.7th). Values from Ref. 571. C: examples of organs, tissues, and cell types that contribute to and determine V̇o2max. Figure created with BioRender.com.


o2max integrates functional aspects of a number of organs and tissues that contribute to oxygen intake, distribution, extraction, and usage (FIGURE 5C) (598). Intake is affected by pulmonary capacity and function, in part depending on respiratory muscle functional capacity, which can be improved by specific training even in older adults (599). Distribution combines cardiac output parameters, oxygen carrying capacity by red blood cells, blood volume, and vascular properties. Extraction and usage, at least in the case of cardiopulmonary exercise tests, are mainly determined by the degree of tissue vascularization (hence the proximity of blood vessels and muscle cells), intramyofibrillar trapping of oxygen by myoglobin, and the rates of mitochondrial oxidative phosphorylation. The rate-limiting step that determines V̇o2max can be variable and, for example, shift depending on the training state, from oxygen usage in muscle fibers in the untrained to oxygen provisioning by cardiac output and tissue vascularization in trained athletes.

6.1.2. V̇o2max: age dependence and health/mortality prediction.

o2max decreases progressively with age, at a rate of ∼7–10% per decade (corresponding to 4–4.6 mL/min/kg) (571, 600, 601), down to an “aerobic frailty threshold” of 17.5–18.0 mL/min/kg that is required for an independent lifestyle (574, 602, 603). At this point, individuals have to utilize almost their maximum aerobic capacity for tasks related to daily life and independence, associated with severe physical fatigability (604). In the worst case, this deterioration continues to fall below 10.5 mL/min/kg, when ∼30% of oxygen is used to maintain basal metabolic rate, potentially leading to fatal outcomes (574, 602). Accordingly, the assessment of V̇o2max constitutes a strong and independent predictor of morbidity and mortality in different populations, young and old, healthy and clinical (574, 576, 605). In fact, this strong link with mortality and various chronic conditions including heart failure, hypertension, stroke, chronic kidney disease, dementia, and depression was consistently demonstrated in an overview of meta-analyses that included >20.9 million observations (576). More specifically, having a low cardiorespiratory fitness is associated with a 41–53% higher relative risk for all-cause mortality compared to those with a high cardiorespiratory fitness (606). Strikingly, even in unfavorable conditions, such as abnormal glycemic status (607) or obesity, being fit is a strong predictor for reduced all-cause and cardiovascular disease mortality (608611). In fact, relatively fit obese men (top 80% of the age-specific cardiorespiratory fitness) have a 50% lower cardiovascular disease mortality risk compared to normal-weight unfit men (lowest 20% of the age-specific cardiorespiratory fitness) (609). Indeed, cardiorespiratory fitness can mitigate risks of obesity in a significant manner, beyond those of unfit, normal-weight individuals (612). Similarly, high cardiorespiratory fitness can overcome an unfavorable genetic predisposition for dementia (613). Furthermore, cardiorespiratory fitness is a strong inverse predictor of heart failure risk irrespective of BMI (614). Importantly, even though up to 50% of the variability in V̇o2max in sedentary individuals is estimated to be of genetic origin (615) as well as other biological and methodological factors (616), trajectories can be strongly affected by exercise at young and old age (617622), and even a moderate increase by 3.5 mL/min/kg [1 metabolic equivalent of task (MET)], achievable after 2–3 mo of training (602), reduces the risk of heart failure by 18% (572) and all-cause mortality by 11–17% (576, 606). Interestingly, despite exhibiting lower V̇o2max values, older athletes can reach high performance levels due to the ability to perform work closer to the V̇o2max compared to younger counterparts (623). Lifelong exercise habits provide most benefits to mitigate age-related declines in V̇o2max (624) and can confer benefits in old age (625). However, even in the very old, cardiorespiratory fitness can be improved, as exemplified in the case study of a 101-yr-old cyclist (626).

6.1.3. V̇o2max: additional tests, benefits, and alternative assessment methods.

A cardiopulmonary exercise test can provide additional health-relevant parameters and can easily be expanded (589592, 627). For example, this test often includes a measurement of the rate of carbon dioxide production (V̇co2). The respiratory exchange ratio (RER: V̇co2 divided by V̇o2) indicates fuel utilization, from oxidation of fatty acids (RER ∼ 0.7) to glucose (RER = 1.0). Thereby, the metabolic preference and transition during a cardiopulmonary exercise test can be assessed and the switch to anaerobic metabolism observed when the RER exceeds 1, sometimes used as exhaustion end point criterion. The RER, heart rate, minute ventilation [V̇e (composed of the tidal volume multiplied by the breathing frequency)], or work rate can be set into relationship to V̇o2 and V̇co2, thereby providing insights into the relative efficiencies and potential deficiencies of the cardiopulmonary system, e.g., by calculating the ventilatory equivalents VEqO2 and VEqVCO2 (corresponding to the ventilation required to take up or exhale a given amount of O2 and CO2, respectively, by dividing V̇e by V̇o2 or V̇co2). Additional health-relevant parameters are obtained from simultaneous (and/or postexercise) acquisitions of stress electrocardiograms (628, 629), blood pressure (630, 631), pulse oximetry or arterial blood gas measurements (632), cardiac magnetic resonance imaging (633), heart rate response and recovery (634636), heart rate variability (637, 638), invasive cardiopulmonary hemodynamics with a pulmonary artery catheter in the internal jugular vein (639), or other parameters. Furthermore, timed blood sampling and determination of lactate levels help to pinpoint the lactate threshold or, when combined with V̇co2 measurement, the anaerobic threshold, even though this concept remains somewhat contentious (640, 641). Outside of clinical testing, V̇o2max correlates with endurance exercise capacity but itself is only one of the key parameters besides fractional utilization of V̇o2max (related to the individual lactate threshold and critical power), exercise economy, and physiological resilience to determine performance (642644). Optimally, all four are measured and optimized in athletic training. It is important to point out that many of the clinically used techniques, parameters, and readouts have emerged from the study of elite athletes (645), who, even though few in number (646), represent the upper limit of human performance capabilities (295, 647). Of note, various protocols exist to estimate V̇o2max in other settings, including submaximal efforts, from maximal and resting heart rates, or based on the Fick equation with cardiac output and the arteriovenous oxygen difference as parameters, e.g., in health-compromised individuals. Therefore, readouts for cardiorespiratory fitness can also be acquired without measuring actual V̇o2max (648). For example, time on treadmill, peak speed, incline, work performed, and other parameters have been used to assess cardiorespiratory fitness, showing good correlation with measured maximal oxygen uptake and with mortality risks (649657). Other parameters such as sex, age, BMI, waist circumference, resting heart rate, physical activity levels, and smoking status can be used to approximate the state of cardiorespiratory fitness (658, 659). In fact, >28 equations have been proposed, using overlapping and distinct metrics, all of which significantly correlate to measured cardiorespiratory fitness, albeit with differences in accuracy (660). Even though measured cardiorespiratory fitness provides better discriminative ability, estimated cardiorespiratory fitness is a valid indicator of health status and mortality risks (659, 661663). For example, activity-induced oxygen uptake, expressed as MET (3.5 mL O2/min/kg), can be calculated with the heart rate index (the ratio of maximal to resting heart rate during and before an activity, respectively) [METs = (6 × heart rate index) − 5] (582, 664).

6.2. Relative Lean/Muscle Mass

6.2.1. Body mass index and the obesity paradox.

Historically, even though controversial (665), an “obesity paradox” has been reported, indicating reduced mortality of patients with an elevated BMI (expressed in kg body mass/m2 height) beyond the normal/healthy range (666). Different explanations have been put forward for this observation, for example based on data indicating that when measures of fat depot distribution are used the obesity paradox disappears (667). Since such measures are independent of differences in muscle mass, they support the muscle mass hypothesis, suggesting that only those patients for whom the elevation in BMI is to a significant extent caused by higher muscle mass experience the health benefits, at least in certain pathologies and populations (666, 668671). The BMI thus is an imperfect marker for obesity (672). Indeed, the relative amount of lean or, better, actual skeletal muscle mass (often normalized to body mass) is a much better predictor of all-cause and disease-specific mortality (673684). Muscle mass is of particular significance in aging (sarcopenia) and cancer (cachexia), with strong correlations to functional capacity, quality of life, morbidity, and survival (685693).

6.2.2. Relative lean/muscle mass: methods.

In contrast to the BMI, which can easily be measured with a scale and a measuring tape, determination of the vastly more meaningful body composition requires specialized equipment, with variances in preciseness and accuracy (694704). Moreover, these methods have different capabilities in determining compartments such as adipose tissue-free mass (ATFM; body mass − adipose tissue mass), fat-free mass [FFM; residual mass (organs such as liver, pancreas, etc.) + fat-free skeletal muscle mass + bone mass], lean soft tissue [LST; residual mass (organs such as liver, pancreas, etc.) + fat-free skeletal muscle mass], or actual skeletal muscle mass, which contributes ∼45–50% to fat-free mass (705). Sometimes, compartments are normalized to other anthropometric measures, e.g., in the fat-free mass index (FFMI), which is indexed to height (expressed in kg FFM/m2). Commonly used methods to measure muscle mass or surrogates thereof include (706710) skinfold measurements with a measuring caliper, in which body composition is inferred from the thickness of subcutaneous fat in different body areas; circumference of various muscles such as mid-upper arm or calf; bioelectrical impedance analysis (BIA), recording the electrical resistance with multiple electrodes and in multifrequency measurements in a body segment-separated manner; dual-energy X-ray absorptiometry (DEXA), in which spectral imaging using two X-ray beams with different energy levels allows the acquisition of body composition data in total body and regional segments (including assessment of bone mass and mineral density); ultrasound, applied to multiple body regions similar to the skinfold calipers; quantitative magnetic resonance (QMR) or magnetic resonance imaging (MRI), in which magnetic fields are used to quantify, and in the case of MRI also visualize, fat and lean mass independent of hydration status; hydrostatic weighing or air displacement plethysmography, using object displacement of water and air, respectively, to calculate body density and subsequently composition; and computed tomography (CT), in which multiple X-ray measurements are processed for a tomographic reconstruction of a body, sometimes combined with positron emission tomography (PET) to assess metabolic activity such as glucose metabolism. Finally, in deuterated creatine dilution, skeletal muscle mass is determined noninvasively, since almost 98% of the creatine pool is found in muscle, colocalizing with sarcomeric structures, which are the functional components of muscle (711713). Therefore, skeletal muscle mass measured by this method strongly correlates with strength, functional capacity, and mortality risks (714, 715). As in all areas of medical diagnosis (716), the use of machine learning techniques and artificial intelligence might help to improve the predictive strength of imaging-based techniques for body composition in the future (717). Moreover, improvements and/or the acquisition of additional parameters will further boost the use. For example, the phase angle [PA; angular transformation of the ratio of capacitance (Xc) to resistance (R): arctan (Xc/R) × 180°/π] measured at 50 Hz in BIA is an indicator of cell health and function and correlates with disease risk and mortality (718720). However, as with most of these methods, better standardization and more normative data for different instruments and populations will help to improve this measure (721723).

6.2.3. Relative lean/muscle mass: adipose tissue content and distribution.

Of note, costly bespoke determinations can sometimes be circumvented by leveraging other, already existing radiological images, e.g., to estimate temporalis muscle thickness in cranial imaging as a marker for muscle mass predictive of disability and mortality (724). As an added benefit of some of these methods, information on adipose tissue distribution (e.g., abdominal vs. subcutaneous) or even fat content of different organs (725) [including myosteatosis (726) or intermuscular adipose tissue (727)] is acquired, both of which are linked to pathological processes in cardio-metabolic diseases (728) and all-cause and cause-specific mortality (729). For example, the determined values of the adipose-free muscle volume and the percentage of intramuscular fat in the thigh muscle by MRI can be compared to reference values and provide predictive data on health (730) or, at least in certain populations, on brain volume (731). Adipose distribution, however, can also be estimated by the much cheaper and simpler waist-to-hip circumference ratio, with good correlation to morbidity and mortality risks (732), which can develop in different trajectories compared to the BMI (733). Other parameters that should improve on the use of BMI have been proposed, including the weight-adjusted waist circumference index (WWI; calculated by dividing waist circumference by the square root of body mass) (734, 735), the body roundness index {BRI; calculated as 364.2 − 365.5 × √(1 − [waist circumference in cm /2π]2 / [0.5 × height in cm]2)} (736), or height-normalized abdominal body composition (737). The combination of muscle strength reduction and fat mass gain, as observed in sarcopenic obesity, might constitute an additional burden on health and mortality (738, 739), for example for the risk of dementia (740). Regardless of the method used, it is undisputed that strong associations of body composition with aging and health exist (741, 742), e.g., in terms of risk for frailty in sarcopenic obese and presarcopenic individuals (743), in health and pathologies, for example nonalcoholic fatty liver disease (NAFLD) patients (744, 745). Notably, the genetic contributions to muscle mass and function are estimated at 30–50%, implying that a majority of the variations are modifiable by environmental factors (746). Nevertheless, despite the recent arrival of new antiobesity drugs, rising rates in overweight and obesity are observed worldwide, accompanied by the corresponding obesity-related pathologies, necessitating effective and aggressive measures targeting environmental and lifestyle factors (747).

6.3. Muscle Strength and Power

6.3.1. Muscle force trumps mass.

In recent years, it has become clear that an exclusive focus on muscle mass is insufficient to describe sarcopenia (muscle wasting in the aging process) and other diseases (748). In fact, the term dynapenia (or powerpenia) has been proposed to describe the functional loss of skeletal muscle, which can be dissociated from changes in body or muscle mass (749752). In aging, the loss in power is greater than that in strength, and both are disproportionally larger than the reduction of muscle mass (753755). Chronologically, the decline in power, strength, and mass is accordingly found in this order (755). It therefore is no surprise that decrease in muscle quality, a measure of strength or functionality relative to muscle mass, strongly correlates with mortality and health in aging (756758). As a consequence, therapies that only ameliorate muscle mass, but not functional aspects, will most likely be suboptimal (759761). Indeed, muscle strength [maximal force, expressed in N, sometimes reported as torque expressed in Nm to describe force application for rotational movement of a joint (762770)] and power [scalar product of the vectors of force and shortening/lengthening velocity of the muscle (771, 772), revealing force production over time or speed of force production, expressed in W (773775)] have been closely associated in an inverse manner with all-cause mortality (677, 750, 776780) not only in healthy individuals but also in patients such as those suffering from diabetes (781).

6.3.2. Methods to determine muscle strength and power: handgrip and more.

The determination of handgrip strength with corresponding dynamometers is one of the most common methods, facilitated by the ease of use and the availability of ample normative data (FIGURE 6), for which values are often expressed in relation to body height and mass, handedness, and other anthropometric measures, with strong correlation with future morbidity and mortality (782791) as well as potentially a number of sociodemographic, anthropometric, behavioral, and psychological factors (792).

FIGURE 6.

FIGURE 6.Physiological biomarker grip strength. Age trajectories of grip strength in men and women. Grip strength data from Ref. 782 (n = 49,964 participants; men: n = 23,277; women: n = 26,687). Adjusted hazard risk ratios from Refs. 783, 784. Figure created with BioRender.com.


Intriguingly, handgrip strength has predictive power for processes beyond muscle status, e.g., extending to frailty (793), glycemic measures (794), hypertension (795), brain (791, 796) and cardiovascular health (797), bone mineral density in women (798), and self-assessed quality of life (799), and associates with a wide variety of anthropometric, morphological, and functional factors (800) and the risk for falls (801). The values obtained with handgrip dynamometers can be approximated with simple tests such as bottle opening or newspaper tear-off in old populations, albeit in the absence of broad, normative datasets (802). However, the measurement of a single strength component might be insufficient to capture more complex or different tasks, for example those related to activities that involve both upper and lower body functions (803, 804). Therefore, specialized, mostly isokinetic dynamometric equipment is used to measure the strength or power of single-joint movements (e.g., knee extension) or multijoint and multimuscle involvement such as acquired with mechanographical force/power plates in plyometric exercises. Furthermore, standard isotonic gym equipment can be leveraged to assess parameters such as the one-repetition maximum (1RM), maximum voluntary isometric contraction (MVIC), muscular endurance (isometric or velocity loss with increasing number of repetitions), dynamic strength index (bar velocity related to force production), reactive strength index (RSI; divide drop jump height by ground contact time), and others. In a frail population, sit-to-stand tests (for example, timing of 5 repetitions of sit-to-stand-to-sit movements without the use of arms and hands or number of sit-to-stand repetitions in a 30-s window) or similar interventions might already be sufficient to estimate strength of the trunk and lower extremities. These measurements can be combined with electromyography (EMG) to obtain additional information on neuronal activation, action potential transmission, motor unit recruitment, and fatigue (805, 806). Moreover, clinical relevance can be expanded by the utilization of tensiomyography (speed of muscle contraction under isometric conditions) and myotonometry (measurement of reaction to a short mechanical impulse), providing data on muscle composition, architecture, and viscoelastic properties (e.g., muscle tone, stiffness, and elasticity), respectively (807). Such approaches might be complemented and expanded with wearable superresolution myographic sensors in the future (808). Of note, integrated approaches, for example assessment of muscle mass and strength, potentially combined with other parameters such as a nutritional score, might increase the predictive power, e.g., for cancer mortality (809).

6.4. Step Count, Leisure-Time Physical Activity, and Sedentary Behavior

6.4.1. Occupational and leisure-time physical activity in the modern world.

The evolution of humankind as persistent hunters is not reflected in the engagement in physical activity in modern societies, with various detrimental consequences for health (264, 295). The amount of physical activity is profoundly different between professions, which can result in beneficial outcomes, e.g., the lower incidence of coronary heart disease in conductors, constantly climbing and descending stairs, compared to the sedentary drivers of double-decker buses in London (810813). Overall, ∼80% of jobs in the United States are estimated to be predominantly sedentary (814).

Discrepancies also exist for nonoccupational leisure-time activity, and a big proportion of the general population do not meet the current WHO guidelines recommending at least 150–300 min/week of moderate aerobic activity and regular muscle-strengthening exercises (815820). For example, only 22.8% of 2,629,508 adults adhered to muscle-strengthening exercise guidelines (821) and less than 52%, 35%, and 28% of the general United States population met endurance, resistance, and combined endurance and resistance training recommendations, respectively, in the year 2018 (822). Reaching the activity levels defined by these guidelines confers substantial health benefits and lowers mortality risks (823826), e.g., up to 31% on all-cause mortality in elderly individuals (827), independent of cardiorespiratory fitness (828, 829). Importantly, even relatively small shifts toward physical activity behavior can elicit beneficial effects (830), which can extend to nonmuscle tissues and functions, e.g., cognition in acutely hospitalized older adults (831) or cancer (832). In fact, all aspects of resilience (833) and intrinsic capacity (834, 835), comprising locomotor (836), cognitive (837), psychological, sensory, and vitality capacity (838), are positively affected by physical activity (839). However, although positive outcomes are already seen at lower doses, additional value can be achieved with higher intensity/volume/frequency (840), albeit with diminishing returns (841843). Inversely, a negative association of leisure-time, nonoccupational physical activity to morbidity as well as mortality has been demonstrated (844856). For example, a strong correlation between leisure-time physical activity and the risk for a number of cancers has been found in large cohorts (857860), with a potential involvement of the exercise-remodeled immune system in this context (861). Importantly, the impact of physical activity is seen regardless of the time of day of performance (862), indicating that activity can mitigate potential adverse effects of circadian timing (863). Similarly, physical activity can overcome other markers of low functionality, e.g., ameliorating mortality risks in individuals with low handgrip strength or preventing cardiovascular disease incidents across all handgrip strength levels (864).

Of note, sedentary behavior (as in time sitting), which is on the rise in many societies, should be assessed independently from other forms of inactivity because of the marked negative effects of time sitting on health and disease risk that can be distinct from those arising from minimal physical activity (865868). Accordingly, sedentary behavior can increase mortality risks, e.g., those for all-cause and cardiovascular diseases (869873), which at least in part can be blunted by physical activity, in particular when performed at high intensity (874880).

6.4.2. Methods to measure leisure-time physical activity: from step count to more sophisticated wearables.

Leisure-time physical activity and sedentary behavior data are often collected by self-reporting, with the corresponding limitations on data accuracy and standardization. More recently, advances in the use of wearable devices have helped to acquire such behavioral data in a more objective and quantifiable manner and have confirmed the positive effect of physical activity on mortality risks (865, 880884). Daily step count is the simplest parameter for which a good association with morbidity and mortality has been demonstrated (885894), e.g., on incident risk of dementia (895) or of depression (896). More sophisticated accelerometers, however, allow a detailed and fine-grained acquisition of different types of activity (or sedentary behavior) in relationship to health and mortality (865, 897899). For example, the personalized activity intelligence score assesses the cumulative fluctuations of heart rate of the most recent 7 days as a measure of relative intensity and energy expenditure of weekly physical activity, shown to correlate with mortality risks (900, 901). Such refined manners of acquisition of behaviors are of particular importance to quantify time sitting (902). At the moment, wearable-based assessment of physical activity is widely deployed to capture endurance training types of activities, using actigraphy, accelerometry, GPS tracking, and heart rate measurements besides other sensors, even though issues with heterogeneity, accuracy, and standardization still exist (903). However, strong health and mortality benefits also arise from resistance training (904920), even at lower intensities/volumes (921), for which corresponding wearables that objectively and accurately quantify the work performed and actual (nonresting) exercise time are still under development (922). In fact, muscle strength and cardiorespiratory fitness are independent predictors of mortality, with best outcomes when performed in combination (923926).

6.5. Gait Speed/Frailty Parameters

6.5.1. Neurodegeneration and sarcopenia are the major drivers of loss of independence, morbidity, and mortality.

In the absence of any debilitating disease, neurodegeneration (927, 928) and sarcopenia (257, 929932) are two of the main factors that precipitate loss of independence, decreased quality of life, morbidity, and mortality in aging (933). The effects caused by these two processes are exacerbated by compromised cardiovascular function (934937) and loss of bone mass and mineral density (938). Osteopenia/osteoporosis is a prominent issue in postmenopausal women (939) but should not be overlooked in men (940). Of note, bone mineral density correlates with lean body mass and muscle strength (746) and can be improved by exercise, in particular in combination with the intake of dairy products (941). Similarly, neurodegeneration and sarcopenia (and inversely physical activity) are mutually linked (942, 943), e.g., in the reduction in vestibular and proprioceptive abilities, leading to altered gait and decreased senses of balance and motor coordination. With the progressive loss of the functional capacity caused by these events, a vicious cycle is initiated and fueled (944), in which insecurities in gait and balance reduce the drive for physical activity (and social interactions), which, in turn, accelerates neuronal and muscle degeneration (FIGURE 7). In the worst case, falls occur (945), with the risk of fractures, immobilization, and hospitalization, further promoting this cycle through a “catabolic crisis” (946949), ultimately culminating in a broad hospital-associated deconditioning (950) and hospital-acquired complications (951).

FIGURE 7.

FIGURE 7.Age-associated degeneration of skeletal muscle, neuronal tissue, bone, and the cardiovascular system drive a vicious cycle leading to loss of independence, morbidity, and mortality. Sarcopenia (loss of muscle mass and function) together with reduced cardiovascular and respiratory function (leading to decreased endurance and increased fatigability) reduce physical performance and increase the perception of effort for exercise- and daily task-related endeavors. As a consequence, such activities are increasingly avoided, further depleting functional capacity and reserves. The lack of adequate levels of these leads to frailty, which, in turn exacerbates sarcopenia, cardiovascular dysfunction, neurodegeneration, and osteoporosis, thereby fueling a vicious cycle. Together with neurodegenerative events, e.g. linked to dementia, this constitutes the major driver for the inability to perform daily tasks (e.g. carrying groceries, cleaning the apartment, walking up and down stairs or across pedestrian crossings), enjoy social interactions, and live independently, thus leading to admission to nursing homes, increasing the risk for (co)morbidities and elevating mortality risks. Neuromuscular deterioration, e.g., in balance, motor coordination, and gait, promotes insecurity and, exacerbated by muscle weakness, also the risk for incident falls, which, in the worst case, lead to fractures (facilitated by reduced bone mass and mineral density), immobilization, and hospitalization. All of these factors contribute to the avoidance of physical activity and exercise, thus further fueling this vicious cycle. Figure created with BioRender.com.


6.5.2. Gait speed and other tests for frailty and neuromuscular functionality.

Based on the deterioration of the neuromuscular system, there is little surprise that functional capacity parameters depending on neuromuscular functionality are predictors of an independent lifestyle, morbidity, and mortality, as seen in the example of gait speed (282, 952960). Gait speed, representing voluntary locomotion, can easily be assessed with a stop watch, pedometers, or accelerometers (961). Additional information can be derived from a more sophisticated analysis of gait, in which for example speed, cadence, stride length, step width, and other parameters describing footprint and gait dynamics are acquired and collectively allow a more comprehensive assessment of aging-related alterations in gait (962, 963). Such data can be obtained with different methods, including pressure measurements, motion capture, or wearable sensors (964). Gait speed trials often are combined with other tests of frailty, for example, cognitive and sensory function, psychological and social aspects, balance and motor coordination, mobility and flexibility, muscle strength and endurance, muscle and body mass loss, fatigability/exhaustion, and integrated tasks of gross and fine motor skills (965968). Such compound frailty assessments are predictive of mortality as well as incident disability, falls, hospitalization, and health care dependence (969972), even in long-term trajectories (973). For example, strength, balance, and gait speed can be used to predict the risk of incident dementia (966, 974). However, even simple tests such as sit-to-stand time (975), sit and rise from the floor (976), or 10-s one-legged stance performance (977) can be used to estimate frailty, functional capacity, mortality, and survival. Stair descent phenotypes can reveal deficits in balance, coordination, muscular agility, and strength and thereby help to predict the risk of incident falls (978). A myriad of test batteries and protocols have been established, aiming for test coverage of a broad and representative range of frailty, impinging on vulnerability to adverse events, reduced resilience toward stressors, and loss of functional capacity (955, 969, 970, 979), all of which clearly associate with the process of (advanced) aging (980, 981).

7. CLASSIFICATION OF BIOMARKERS OF AGING

The aforementioned physiological parameters belong to a group of biomarkers of aging, some of which, e.g., the 6-min walk test, have already been accepted by the FDA as surrogate in clinical trials (570). In terms of application, physiological biomarkers fulfill the criteria of multiple categories (506):

1) 

Predictive biomarkers: The physiological biomarkers have a strong and independent predictive power for all-cause mortality and various disease-specific risks.

2) 

Prognostic biomarkers: For most of the functional/physiological biomarkers, predictive power is not only limited to healthy individuals but extends to patients in very diverse settings, for example as a measure for fitness for and outcome of surgery (982987), clinical outcomes in heart failure (988), coronary bypass grafting (989), intensive care unit hospitalization (990, 991), frailty (970, 992995), cancer mortality (996, 997), sarcopenia (998), biliary sepsis (999), liver transplantation (1000), hospitalization secondary to COVID-19 or other infectious diseases (1001), potentially linked to reduced respiratory function (1002), or cognitive impairment and dementia (1003). Notably, the corresponding interventions, in particular exercise training, can be used in a prognostic manner in prehabilitation to mitigate loss of muscle mass and function, increase resilience, reduce adverse outcomes, and shorten the duration of hospital stays (1004).

3) 

Response biomarkers: The physiological biomarkers not only predict morbidity and mortality but also react to interventions that improve prospects, first and foremost physical activity, the most robust intervention known to date to promote healthy aging.

4) 

Surrogate end point markers: Because of the extraordinary correlation between physiological biomarkers, biological age, morbidity, and mortality, interventions aimed at the aging process should be benchmarked against these measures, whenever possible in a comprehensive manner. For example, weight loss caused by caloric restriction is not expected to change absolute V̇o2max, even though oxygen consumption normalized to body mass can increase or could lead to a reduction in lean body mass if performed in the absence of concomitant resistance training (1005, 1006). However, leisure-time activity, number of steps, as well as gait speed and other frailty markers could improve, at least based on extrapolation of data from mice.

5) 

Discovery biomarkers: Physiological biomarkers describe the integrated function of various tissues, organs, and cell types and thus reflect the multifactorial processes and complexity of aging. However, the underlying mechanistic principles are still poorly understood and therefore harbor an enormous potential to reveal novel insights into the benefits of interventions that improve these biomarkers, as well as the patho-etiology of aging-linked processes.

7.1. Assessment of Biomarkers of Aging

Four main criteria for ideal biomarkers of aging have been put forward (506). The physiological biomarkers, in particular in combination, fulfill all of these:

1) 

Measurement: The assessment of all of the physiological biomarkers is minimally invasive, and, maybe with the exception of self-reporting-based values, highly reliable. Therefore, longitudinal assessment, lifelong, from young to very old age, is feasible.

2) 

Aging relevance: The physiological biomarkers not only predict morbidity and mortality but also provide a snapshot on functional capacity, resilience, and (organ) health.

3) 

Predictive power for functional aspects of aging: This is clearly provided by the physiological biomarkers, extending to practical and tangible aspects in daily life, e.g., impaired gait speed resulting in the inability to cross roads (1007).

4) 

Responsiveness to longevity interventions: All of the physiological biomarkers are pliable and thus responsive to interventions, in particular those with most benefits on the aging process (1008).

7.1.1. Feasibility and validity.

The determination of physiological biomarkers, including V̇o2max, muscle mass and strength/power, gait speed, locomotor activity, and frailty, is minimally invasive and nonlethal in model organisms, at least in higher vertebrates. A longitudinal assessment therefore is possible, even desirable, to monitor trajectories over time, facilitated by the short time and ease of acquisition. Importantly, these measurements are non-age-accelerating; in fact, the tests, at least in some cases like V̇o2max, muscle strength/power, and gait speed, represent the intervention and therefore contribute to the beneficial effects. Then, physiological biomarkers are age sensitive, with high correlation with chronological age. However, in contrast to other molecular biomarkers for which accuracy for chronological age seems to come at the expense of predictive power for mortality and age-associated health outcomes, physiological biomarkers have been demonstrated repeatedly to provide very accurate prediction of morbidity and mortality, and possibly “biological age” (FIGURE 8).

FIGURE 8.

FIGURE 8.Physiological aging biomarkers associate with chronological age, while also predicting “biological age.” In this figure, the decline of gait speed with chronological age in men and women is shown from which a biological age can be inferred. In the examples, the green star represents an individual of chronological age 80 yr, who, however, has a gait speed representing the mean of 70 yr of age. In contrast, the slow gait speed of the 70-yr-old individual (red star) is equivalent to that of 80-yr-old individuals. In these examples, the 80-yr-old individual would still sequester above the threshold of 1.2 m/s gait speed needed to walk across many pedestrian crossings (indicated by the yellow line) (see Ref. 1009), whereas the 70-yr-old individual would fail to succeed in this task. The figure is based on data presented in Ref. 1010 (men: n = 2,087; women: n = 2,569). Figure created with BioRender.com.


Importantly, in contrast to most contemporary molecular biomarkers, extensive epidemiological, prospective, longitudinal, and cross-sectional data in humans exist for the physiological biomarkers in that regard. Thus, age-sensitive criteria are fulfilled: good prediction of all-cause mortality as well as correlation with multiple age-sensitive features, i.e., age-associated morbidities, by providing information about functional aspects of multiple systems, integrating different signals, and incorporating heterogeneous aspects. Two types of information are provided, integrity and resilience of tissue/organ function as well as, in the case of longitudinal assessment, rates of progression of deterioration (or mitigation/reversion by interventions).

7.1.2. Mechanistic criteria and biologic plausibility.

In the absence of confounding age-associated diseases, inevitable neurodegeneration, sarcopenia, and functional decline in the cardiovascular system are the main drivers for elderly individuals to lose independence, be admitted to nursing homes, and experience increased morbidity and mortality (1011). Collectively, these processes promote an inactive lifestyle due to increased perception of effort and insecurities (e.g., related to a decline in balance and motor coordination), leading to a vicious, self-reinforcing cycle. In the worst case, elderly individuals fall, and the ensuing fracture, facilitated by osteopenia/osteoporosis (in osteosarcopenia) (1012) and potentially exacerbated by osteoarthritis (in musculoskeletal failure) (1013), leads to immobilization and hospitalization. Inversely, an active lifestyle, in particular when enriched by endurance, resistance, balance/agility, and flexibility training (1014), is the best, indeed so far only, intervention to mitigate sarcopenia and neurodegeneration (1015) and one of the best to counteract osteopenia/osteoporosis and boost cardiovascular function. In fact, endurance, strength, and flexibility training have all been shown to improve mortality risks (1016, 1017). Fall risks are reduced by strength and balance training affecting posture, gait, and coordination, resistance training improving sarcopenia and joint mobility, and cognitive exercises boosting spatial awareness and attention (1018). Thus, a clear antiaging effect has been demonstrated, impinging on various deleterious processes that affect health span and lifespan.

The aforementioned physiological biomarkers exhibit a well-described deterioration with “biological age,” e.g., in terms of muscle mass and strength, cardiorespiratory function, gait speed, or frailty. Whereas the latter are primarily affected in old age, the former exhibit an association starting at younger ages. Thus, even though the pathways and molecular underpinnings of, and the potential health benefits elicited by, interventions aimed at these biomarkers are still only poorly understood, a strong biological plausibility exists that links functional resilience of the cardiovascular and neuromuscular systems to aging, morbidity, and mortality, and most likely impinge on fundamental aspects of aging. Of note, the effects of the interventions that are directly related to these biomarkers transcend health and function of the primary target tissues. Thus, exercise-based interventions not only improve muscle and cardiovascular function but affect almost every organ and system in the human body in a clinically relevant manner (296, 302, 1019, 1020). For example, muscle mass and strength as well as the amount of physical activity are negatively associated with the relative risk for dementia and a decline in cognitive function, brain structure, neurodegeneration, and mental health (1003, 10211032), even when performed in an irregular manner, e.g., by “weekend warriors” (942, 1028, 10331035). Such activity patterns, concentrated at 1 or 2 days per week, also confer health benefits in other domains, for example incident cardiovascular disease rates (1036). More regular exercise-based interventions obviously also mitigate these risks (1037), potentially even changing lifelong trajectories if initiated early in life (379, 1003, 10381040). Such youth-specific programs could affect life history and thereby influence health aging (1041, 1042).

7.1.3. Generalizability.

Most of the proposed physiological biomarkers can be assessed in model organisms, and at least some show a remarkable similarity to humans, e.g., pliability of V̇o2max (1043), association of V̇o2max with longevity (1044), and deterioration of balance and gait in mice, amenable to amelioration by exercise (1045), if certain biological and methodological issues are considered (97, 1046). Thus, these biomarkers can be studied mechanistically in multiple species, with a high translatability potential (1047). In humans, importantly, these biomarkers have been validated in different clinical populations and demographics (573, 784, 888, 1048), even across different age groups (573, 773, 1049). For example, cardiorespiratory fitness in youth predicts age-associated diseases at old age, e.g., for site-specific cancer (1050). Similarly, midlife grip strength correlates with functional capacity and resilience at old age (1049) and youth sport participation with sarcopenia (1051). Furthermore, these parameters, and the corresponding interventions, are equally valid from the youngest (1052) to the oldest of the old (766, 953, 954, 10531063), as exemplified by a case study of a 71-yr-old world champion powerlifter who started resistance exercise at the age of 63 yr (1064) or a late bloomer octogenarian triathlete (1065). Of note, benefits are found even in suboptimal conditions, e.g., obesity (608, 1066), Alzheimer’s disease (1067), schizophrenia (1068), poor sleep (351, 1069, 1070), hospitalization (1071), rheumatoid arthritis (1072), hypertension (10731075), pulmonary hypertension (1076, 1077), heart failure (1078) and cardiac rehabilitation (1079), chronic obstructive pulmonary disorder (COPD) (10801082), diabetes (1083), chronic kidney disease (1084, 1085), depression (1086, 1087), and even multimorbidity (1088). At the moment, very few pathological contexts contraindicate the use of physical activity, as for example hotly debated in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), in which patients can experience a postexertional malaise lasting for several days (1089). Nevertheless, physical activity levels are directly correlated with all-cause mortality even in individuals with other risk factors, such as cigarette smoking or early parental death (1090, 1091). Inversely, physiological biomarkers can be uncoupled from the genetic background and accordingly are pliable even between monozygotic twins (10921095). Moreover, consensus is emerging that absolute nonresponders to physical activity do not exist, inasmuch as such individuals might respond to different training paradigms, intensities, or volume or might have been misclassified as nonresponders because of measurement and other technical errors (295, 10961100). This extreme clinical generalizability is different from many of the proposed preclinical interventions aimed at lifespan and/or health span extension, many of which only work in specific mouse strains [e.g., caloric restriction (212)], sex [e.g., the majority of pharmacological approaches (129)], or experimental conditions [e.g., pharmacological approaches (38, 475) or caloric restriction (83, 208210, 219)].

At a glance, some of the existing data on physiological biomarkers seem counterintuitive and suffer from similar drawbacks: for example, even though women have markedly lower V̇o2max (FIGURE 5B) and grip strength (FIGURE 6) than men, the former outlive the latter in most societies in terms of average and maximal life expectancy (1101). Surprisingly, opposite to this improved survival, frailty is more common in women than men, suggesting a sex-based frailty-mortality (or health-survival) paradox (11021107). Biological differences certainly contribute to these observations, e.g., in terms of immune system function or the prevalence of life-threatening versus non-life-threatening chronic conditions (1108). Similarly, considerable sex-based differences in exercise response and performance exist (1109). However, psychosocial, societal, socioeconomic (1110), and educational factors should not be neglected, for example sex differences in the number of doctor visits, inclusion in clinical trials, risk aversion, or engagement in healthy nutritional and other lifestyle behaviors. Indeed, at least in some countries, the survival gap between women and men is narrowing (1111), potentially driven by behavioral changes, socioeconomic factors, and education (1112). Curiously, women also derive greater benefits from equivalent doses of leisure-time physical activity than men, at least in terms of reduction of mortality (1113). Even though this interesting phenomenon still is only rudimentarily understood at the moment, it is important to note that physiological biomarkers predict mortality not only in sex-separated but also in mixed groups. In the future, this predictive power might be further elevated by leveraging group-stratified data or individualized trajectories, making use of a combination of physiological biomarkers that integrate aspects of health, resilience, and deterioration representative of different organs and systems. At the same time, a more personalized approach, based on sex among other factors, in determining functional aspects, health, and well-being, coupled to the design of early and late preventative as well as therapeutic measures, seems necessary.

7.1.4. Response criteria.

The physiological biomarkers of aging reflect accelerated and decelerated aging inasmuch as they accurately predict morbidity and mortality. More importantly, the interventions aimed at these biomarkers, physical activity and exercise, are powerful geroprotectors (37, 38, 264, 924, 1019, 1114).

7.1.5. Cost considerations.

Most molecular biomarkers and clocks rely on invasive sample acquisition, specialized equipment, prohibitive costs, and extensive data analysis, precluding population-wide application, at least at the moment. In contrast, the measurement of most physiological biomarkers is easy and relatively cheap, e.g., step count, grip strength, sit-to-stand tasks, and gait speed. For others, most expense will arise from the initial investments for the acquisition of the corresponding instruments [e.g., gas analyzers for the determination of V̇o2max and dual-energy X-ray absorptiometry (DEXA), bioelectrical impedance analysis (BIA), magnetic resonance imaging (MRI), or computed tomography (CT) instruments for the determination of (segmental) body composition, lean and muscle mass, respectively]. Importantly, these instruments can be reused, even repurposed for additional applications, and require minimal continuing investment. Indeed, large-scale, longitudinal imaging programs with MRI, DEXA, and carotid ultrasound have been successfully initiated with 30,000–100,000 participants (1115, 1116). The notion of cardiorespiratory fitness tests being unduly demanding in resources and costs pales in light of the tremendous significance of predicting health and mortality risks and strongly favors a broad and routine implementation of such tests in clinical practice (11171119). Thus, in general, physiological biomarkers can be cost-effectively collected in large cohorts and longitudinal studies. Moreover, some parameters can even be determined with self-monitoring, e.g., daily steps, gait speed, and sit-to-stand time.

7.1.6. Invasiveness and safety.

In general, the physiological biomarkers can be measured in a low-risk, noninvasive manner. The determination of cardiorespiratory fitness via V̇o2max is the only biomarker that necessitates higher intensities and could thus be more problematic in some cohorts. Standardized guidelines for the determination of cardiorespiratory fitness have been established to implement testing in the clinical setting (11201124). Adequate safety measures should be considered, e.g., preparticipation health screening and the concomitant acquisition of electrocardiographic data and/or blood pressure (11251130). Such measurements provide additional clinical insights into cardiovascular health and potentially masked hypertension. Notably, V̇o2max can also be estimated in submaximal tests (1126, 1131).

Cautionary findings have also been reported for exercise-based interventions, in particular at very high intensities (581, 582, 1132). For example, a U-shaped association between exercise intensity and the occurrence of atrial fibrillation has been reported (1133, 1134), however, diametrically opposed to the clear negative correlation of the risk for atrial fibrillation with cardiorespiratory fitness levels (1135, 1136). A potential “exercise toxicity” could also be inferred from reversed J-type (or U-shaped) mortality curves in other studies, with an increase of the relative risks at very high intensities (11371139). Of note, the confidence intervals for these specific groups are large, because of the low number of participants training at such intensities, and the relative risk still is markedly below that of sedentary individuals (1137). Besides atrial fibrillation, excessive endurance training has also been linked to a higher occurrence of arterial plaques and myocardial fibrosis (1140), even though the association with intensity (1141) or volume (1142) seems complicated (1143) and associations are not seen consistently across studies (1144). For example, a reduction in plaques was seen in an exclusive female Master endurance athlete cohort (1145). At the moment, it is not clear whether these changes are indeed pathological, if they are induced by other factors and risk behaviors (e.g., former smoking habits), or if they represent nonconventional pathophysiology (e.g., calcified vs. noncalcified plaques and/or stabilization of plaques). Indeed, follow-up studies of endurance athletes with increased pathophysiological symptoms revealed no increase in all-cause or cardiovascular mortality (1140), regardless of coronary artery calcification load (1146) or elevated genetic risk for cardiovascular diseases (1147).

In contrast to these cautionary findings, no upper threshold for the mortality benefit of cardiorespiratory fitness was found up to very high levels (571, 573, 1148, 1149). In fact, structural analysis revealed no cardiac changes beyond the normal range, even in individuals with very high engagement in physical activity (1150). Indeed, many studies report “L”-type mortality curves, with no added benefit but also no drawbacks of very high-intensity exercise (1066, 1151). Thus, at some point, increased training load might have a very small additional impact on health parameters (1152, 1153) but, based on most available data, should also not confer pathological outcomes on the cohort level (925, 1154, 1155), even though individuals with unfavorable genetic predisposition and/or morphological, anatomical, or functional abnormalities might be at higher risk (1156). As a case in point, even athletes with enormous training loads, e.g., participants in the Olympic Games or the Tour de France, the first sub-4 minutes per mile male runners, or other former athletes, have better morbidity and mortality scores compared to the general population, or even their noncompeting siblings (295, 11571171). This is irrespective of country of origin, medal, or type of sport (1172) and to a large extent driven by improved cardiovascular and cancer mortality (1158, 1173, 1174). In fact, even epigenetic aging seems decelerated in Olympic champions compared to nonchampions, with hypomethylation of genes involved in synaptic health, glycosylation, metal ion transfer, and force generation as well as hypermethylation of genes associated with cancer promotion (1175). Obviously, none of these effects can be completely dissociated from a selection bias based on other health-beneficial habits that could distinguish this group from their nonelite athlete peers (1176, 1177). However, improved survival is also found in nonelite athletes, e.g., in a study of 546,876 participants of Dutch running, cycling, and walking events that has revealed that even in the short term (7 days), no increase in mortality odds was observed, whereas a 30% lower risk of death ensued in a 3.3-yr follow-up (1178). Benefits most likely persist and could confer lifelong health advantages, e.g., as reported in the case study of a 77-yr-old former world-record-holding marathoner (1179). Importantly, low incidence rates of cardiac arrest were also reported during long-distance events including half-marathon (0.27 per 100,000 participants), marathon (1.01 per 100,000) (1180), and triathlon (1.74 per 100,000) (1181). Indeed, the estimated rates of sudden cardiac death in athletes that range from 1:40,000 to 1:300,000 are much lower than those observed in the general population at 1:2,000 (97, 1182).

In summary, in the absence of adverse pathologies, exercise is an extremely safe intervention with proven benefits and thus should be broadly recommended (97), even in old (1183), frail, or otherwise pathological cohorts when certain measures are taken, for example in people with heart failure (581, 582, 1184, 1185). Thus, appropriate preparticipation screening, design, and monitoring of training programs as well as clinical follow-up of vulnerable populations, including the aforementioned potential cardiovascular events associated with high-intensity/volume endurance exercise in veteran athletes, should help to prevent and mitigate any adverse outcomes (581, 582, 11861193). Thus, for most people, the risks and potential adverse effects of physical activity are dwarfed by the benefits (1189). Importantly, such benefits can even arise from small efforts (11941199) such as walking (12001203) or non-exercise-related activities in daily life (1204) and can be long-lasting when following appropriate protocols, e.g., up to 4 yr in volunteers of retirement age undergoing resistance training (1205). The overall accumulation of time of physical activity seems more important than individual bout lengths (12061208), at least when performed at moderate to vigorous intensity (1209). Notably, however, additional effects can be achieved by higher engagement, e.g., in structured high-intensity interval training (HIIT) compared to moderate-intensity continuous training (MICT) or unstructured activity that meets the national guidelines (1210).

7.2. Validation of Biomarkers of Aging

7.2.1. Analytical validation.

Most physiological biomarkers are quantifiable and easy to measure with well-established methods for data acquisition and interpretation, and their assessment is accurate, reliable, repeatable, and reproducible, e.g., V̇o2max, muscle mass, strength/power, and step count. They circumvent issues of many molecular biomarkers in terms of signal to noise in the differentiation of positive and negative results. Similarly, drawbacks in invasive sample acquisition, preparation, storage, and assay do not exist. However, some of the physiological biomarkers can rely on self-reporting and questionnaires, for example the amount of leisure-time activity, or individual assessment of fatigue and well-being in frailty scores. Importantly, with the increasing use of wearable sensors these drawbacks might be overcome in the future, providing more quantitative, accurate, and reproducible data on these parameters (295, 1211, 1212). For example, physical activity can be precisely measured with accelerometers, and correlation with health benefits has been demonstrated (1204, 12131215). Machine learning-based methods can help in the interpretation, and activity or risk recognition can be based on the wearables data (1216). In fact, the use of wearables extends to the improvement of adherence and compliance to physical activity interventions, with clear, long-term clinical benefits (1213, 1217). Bluetooth low-energy sensors, or similar techniques, could extend the usability to the tracking of indoor location and even social interactions (1218). Obviously, issues of validity, reliability, accuracy, reproducibility, standardization, transparency in used algorithms, data privacy, usage, and ownership will have to be resolved before deployment of such instruments (1219). If successful, such applications would further increase the objective quantitation of physiological biomarkers (12201222).

7.2.2. Clinical validation.

In contrast to most molecular biomarkers, physiological biomarkers have an extended history of clinical validation regarding morbidity and mortality as predictive, prognostic, and response biomarkers in various cohorts and populations. Moreover, at least some of these parameters help in clinical decision-making in different patient populations, e.g., in the prediction of postoperative outcomes (982, 996, 1223). In fact, clinical exercise tests, in particular cardiorespiratory fitness measurements, have been promoted as key tests to stratify patient risk profiles and encourage healthy lifestyle choices (1122). Moreover, the higher the adoption of standardized tests, e.g., for cardiopulmonary exercise testing (1224), the better normative reference values for different ethnicities, sexes, age groups, and healthy versus clinical populations and other demographic parameters will be obtained (12251233). To achieve such ambitious goals, national and global registries and multicenter databases for cardiorespiratory fitness values with sufficient representation of various populations, from pediatric to geriatric, have been proposed, including normative as well as criterion-based standards (1234, 1235). Similar datasets should be acquired for the other physiological biomarkers of health and aging. A number of the proposed physiological biomarkers have already been used in longitudinal aging studies such as the English Longitudinal Study of Ageing (ELSA), Health and Retirement Study (HRS), and Longitudinal Aging Study Amsterdam (LASA). For example, reduced function of lower extremities assessed by gait speed and balance is associated with a two to three times higher risk of incident dementia over 15 yr (974). Furthermore, an improvement in one score in the Short Physical Performance Battery (SPPB), evaluating gait speed, balance, and repeated sit-to-stand, is associated with 8% lower odds of falling over a 14-yr period together with a lower risk for other mobility impairments (1236). Similarly, a one-score increase in the physical performance test (including walking, sit-to-stand, and balance tests) and the gain of 1 kg of handgrip strength reduce the 6-yr fracture risk in men by ∼10% and ∼5%, respectively (1237).

7.2.3. Translation of biomarkers.

For many molecular biomarkers, translation to a clinical setting is hampered by several challenges, of which six key barriers have recently been proposed (135). Here, the current position of the physiological biomarkers is discussed in this framework.

1) 

Data sharing for development and validation: Open and free access to publication and data is a problem that is not unique to the field of biomarkers of aging but extends to all of scientific research. It thus is as imperative for the physiological as for the molecular biomarkers of aging that the FAIR (findable, accessible, interoperable and reusable) principles are followed. For some biomarkers, e.g., cardiorespiratory fitness, such attempts currently are ongoing (1120).

2) 

Relative importance of criteria: Even though evaluation criteria for biomarkers of aging have been proposed (506), the relative importance of these is unclear, and arguments for and against can be formulated. For example, as the first of the eight criteria discussed (135), the correlation of a biomarker with chronological age is important but, if too rigid, might not be modifiable by geroprotectors or other factors that affect age trajectories. Of note, physiological biomarkers exhibit both change with chronological age as well as pliability. Second, strong predictive power of all-cause mortality exists, whereas, at the same time, specific risks for sub- and various clinical populations have been found. Third, some of the tests, e.g., gait speed and other frailty assessments, predict functional capacity primarily in older individuals. Others, however, e.g., V̇o2max, are also applicable and valid in younger subpopulations. Fourth, the risk for many age-related diseases can be assessed from physiological biomarkers and, in many cases, direct causality has been proposed, for example linking suboptimal cardiorespiratory fitness to cardiovascular pathologies. Fifth, the physiological biomarkers reflect causal aspects of aging, represented by the universal decline in muscle, neuronal, and bone tissue mass and function. Sixth, the response to factors that accelerate aging, or, better, increase the risk of morbidities and mortality, is given, at least for those with a clear effect such as a sedentary lifestyle (305). Seventh, inversely, the proven geroprotectors, e.g., physical activity, have likewise a positive effect on physiological biomarkers. Finally, the physiological biomarkers have been tested and validated in large and diverse populations.

3) 

Age range for application: With the exception of frailty markers mainly relevant in geriatric populations, physiological biomarkers can be assessed and longitudinally monitored starting at young age and can help to reveal healthy or unhealthy trajectories that are central for the aging process. Importantly, the determination of these factors is safe and noninvasive.

4) 

Minimal criteria for clinical use and implementation: As the physiological biomarkers are already used in clinical practice, such criteria have been met.

5) 

Positioning of biomarkers of aging in the current disease-specific health care setting: Physiological biomarkers are being used for patient stratification, treatment monitoring, disease prevention, and targeted interventions, thus providing clear actionable insights.

6) 

Connecting biomarkers of aging with actionable insights in health care and preventative settings: Physiological biomarkers are excellent measures for individual health monitoring. However, only some are easily implementable on an individual level that does not require access to specialized equipment and facilities. A subset such as step counters and other wearables can be, and are, already widely used, with meaningful outcomes for health and mortality.

In summary, the physiological biomarkers of aging have already overcome most of the challenges that are faced by new molecular biomarkers for clinical translation. Nevertheless, further improvements are still desirable, including open science principles and accessible and affordable infrastructure for the longitudinal monitoring of large populations.

8. CHALLENGES AND PERSPECTIVES

The validity of physiological biomarkers to predict morbidity and mortality in the human aging process is well established. Similarly, the effects of exercise as geroprotector, and as highly efficacious intervention for the prevention and treatment of many pathologies, most of which are chronic and age associated in nature, are undisputed, as is the acceptance of a sedentary lifestyle as a strong and independent risk factor for many different diseases (264, 302, 305). It therefore is mysterious why this knowledge is not leveraged to a greater extent in predicting health, the aging process, morbidity, and mortality in preclinical and human studies. As outlined, the determination of these parameters is noninvasive, non-age-accelerating, easy, precise, and reproducible, in particular with improvements based on wearables data. Moreover, even a widespread screening of large cohorts is relatively cost-efficient once initial investments in instruments have been made.

8.1. A Potential Action Plan for Health Monitoring from Young to Old

A comprehensive, longitudinal assessment of health and aging trajectories would optimally be done in a systematic, multicomponent manner (FIGURE 9A).

FIGURE 9.

FIGURE 9.Comprehensive assessment of physiological and other biomarkers of aging. A: gait speed, grip strength, leisure-time activity, maximal rate of oxygen consumption (V̇o2max), relative muscle mass, and related parameters are clinically proven biomarkers of aging. A longitudinal assessment throughout lifetime could provide information on aging and health trajectories, efficacy of interventions and treatment, and detrimental outcome of pathological events. Data on sleep and nutrition, wearable and/or app-based, could likewise be included. Moreover, specific assessment of vascular function (and “vascular age”), based on blood flow, stiffness, and dilation dynamics, assessed in a noninvasive manner, for example in the retinal vasculature, helps to predict vascular health. Such an individual “health/aging” pass, consisting of a combination of these markers, could further be combined with general health screening, pharmacological preventative interventions (e.g., a polypill), or wearables data, even though the benefits of these measures are currently questionable. In the future, molecular biomarkers of aging as well as potential pharmacological and interventional means could be included in such a strategy, if valid clinical data on efficacy and safety in the human aging process can be shown. B: a personalized health pass covering various domains should be based on the proposed biomarkers and obtained in a longitudinal manner from young to old age. Thereby, favorable or unfavorable trajectories could be identified early on and appropriate measures prescribed. BIA, bioelectric impedance analysis; CT, computed tomography; DEXA, dual-energy X-ray absorptiometry; MRI, magnetic resonance imaging. Figure created with BioRender.com.


Currently, such biomarker assessments have to be stratified by the strength of clinical evidence. Thus, first and foremost, the physiological biomarkers, as discussed in sect. 6, would provide the basis of screening at the present time. The validity might, however, be further amplified by expanding such screenings with other indices of health and comorbidities (1188, 1189), including blood biomarkers for disease risk (11901192) and composite markers for different domains, e.g., endocrine and immune function or cognitive and physical functional capacity (1193). An integration of the physiological biomarkers with the determination of other attributes of functional capacity (1238) (cognition, psychology, hearing, vision, and vitality/nutrition), e.g., used in the WHO Integrated Care for Older People (ICOPE) program to assess intrinsic functional capacity (12391241), reproduced in the INSPIRE animal cohort, could further boost the predictive power and reflect the multidimensional aspects of aging, functional capacity, and resilience (979). Similarly, additional tests of (micro)vascular health, e.g., assessed noninvasively in the retina, could complement V̇o2max data on cardiovascular health and function (1242), maybe combined with other retinal features (1243, 1244). Moreover, wearables-based acquisition of sleep-related parameters (341, 1245) and app-based inventory of nutrition (1246) would cover other important aspects related to the aging process, even provide compositional insights, e.g., on sleep and activity (1247). Improvements in acquisition of such data, in particular in regard to nutrition, would have to be made, since self-reporting, as in other areas, is of limited reliability (334, 335, 1248). At the moment, disease risk and outbreak is mostly monitored with general health screenings, even though the corresponding benefits often are questionable (381). Along the same lines, pharmacological interventions such as broad administration of polypills and some wearables data, for example those claiming to predict atrial fibrillation (1249) and continuous glucose monitoring in nondiabetic patients (1250, 1251), will have to be rigorously tested since clinical use of these in healthy individuals and in aging is still under debate. Little to no data currently exists for proposed pharmacological and interventional “antiaging” strategies as discussed in sects. 2 and 3, as well as the molecular biomarkers of aging. Nevertheless, once positive steps to clinical validation in humans have been achieved, these could also be included in a health screening. Finally, other molecular, morphological, and functional screening tools might be considered and tested in the future, e.g., aimed at estimating immune system function, intestinal or other microbiomes, or blood and urinary biomarkers (1252).

Optimally, a personalized health pass based on such data would be initiated at young age and updated in a longitudinal manner throughout life (FIGURE 9B). Thereby, favorable or unfavorable trajectories could be identified (573, 1050, 1051, 1253, 1254) and appropriate measures initiated in early stages of deterioration. Moreover, the impact of detrimental events, e.g., hospitalization, as well as the benefits of interventions could be quantified. Finally, personalized diagnosis, e.g., of sarcopenia, could overcome prevailing issues with cross-sectional and population data (1255) and might enable early detection and initiation of preventative measures on the individual and/or community levels (1256). Thus, a better integration of these physiological biomarkers in fundamental aging studies in model organisms and humans, population-wide health screening, and clinical trials is one challenge to overcome, even though the hurdles to doing so are low (1257).

8.2. How Can the Adoption of a Healthy Lifestyle Be Increased?

A much larger challenge than the implementation of many of these biomarkers and screenings is the poor compliance and adherence of many people to lifestyle-based interventions despite the obvious health benefits (1258, 1259). In this section, physical activity is used as an example, but analogous conclusions and recommendations could also be made for the better adaptation of other factors discussed in sect. 3. Certainly, various reasons contribute to differences in active and sedentary behavior, including socioeconomic aspects, availability of time, access to facilities, or fatigue (295). However, to a significant extent, human (and murine) activity behavior is controlled by innate, genetic factors (1260). Therefore, vague recommendations to increase leisure-time activity, or to simply overcome lack of willpower, often fall short, even in patient populations with high risks and clear benefit of exercise (1261). In recent years, factors that decisively contribute to adherence and compliance have been studied and quantified (12621268), helping to overcome the intention-behavior gap (1269, 1270). Even the WHO initiated the “Global Action Plan on Physical Activity 2018–2030” with the vision of “more active people for a healthier world,” aiming for a 15% relative reduction in the global prevalence of physical inactivity by 2030 (1271). In many regards, promoting physical activity (and other behaviors with health benefits) can be attributed to four levels (1265, 12721274): political framework, health care systems, health care professionals, and individuals (FIGURE 10).

FIGURE 10.

FIGURE 10.A four-level approach to promote physical activity. Physical activity, exercise training, and calorically controlled, balanced diet have proven benefits on the aging process, morbidity, and mortality. These lifestyle-based behaviors should therefore be promoted on different levels, from a political framework that supports the corresponding infrastructural adaptations to health care system investing in prevention, health care professionals monitoring and prescribing guided, structured programs, and eventually the individual, who will have to initiate and maintain behavioral changes for long-lasting effects. Various examples of measures are shown. Figure created with BioRender.com and Adobe Stock Photos.


8.2.1. Political framework.

For example, the reduction of individualized, motorized vehicles, which are strongly linked to sedentary behavior and unhealthy lifestyles (12751278), in favor of biking, walking, or public transportation and other infrastructural aspects (e.g., central, easily accessible, and attractive staircases instead of elevators and escalators, parks and recreational areas, urban/jungle gyms, walking/running tracks) promotes an active lifestyle. These efforts also include expanding walking and cycling networks and improving pedestrian and cyclist safety (1271). Notably, walkability confers clear benefits on physical activity behavior in healthy individuals (1279) and cancer survivors (1280). Additionally, fostering an active society can involve offering free activities in parks and public open spaces or the temporary/permanent closure of roads to motorized vehicles to facilitate activities such as cycling, inline skating, and walking (1271). Other interventions include reducing adverse behaviors such as usage of tobacco, alcohol, or sugar-sweetened beverages and other unhealthy food and drinks through taxation and the reduction/elimination of subsidies for fossil fuels (to curb anthropomorphic climate change and promote more active ways of mobility), all of which will liberate funds for the promotion of healthy lifestyles (233). The conversion of Paris with the Plan Vélo 2021–2026 shows the feasibility even in large metropolitan areas, given the political will and financial investments. As a side effect, other potentially detrimental factors in healthy aging, e.g., air pollution, noise emissions, and insufficient sun exposure, would also be mitigated by such infrastructural changes. Importantly, research and surveillance of healthy lifestyle habits and interventions should be promoted, as should public awareness.

8.2.2. Health care systems.

A shift of the emphasis from treatment, care, and rehabilitation to prevention (primary and secondary) should be promoted (FIGURE 11).

FIGURE 11.

FIGURE 11.Focus on prevention rather than treatment. A: impaired functional capacity is a major driving force for incident morbidity, accidents, or wear and tear, potentially leading to dependence on medical intervention and hospitalization and the corresponding debilitation. The prevailing model of a reactive, therapy-focused system with high costs and high relapse of rehabilitation is aimed at bringing back patients to the initial baseline of functional capacity, which often remains in a suboptimal range, thus potentially setting up patients for relapses. B: additional programs that boost functional capacity to an optimal level, e.g., with health coaching and guided, structured training programs, could help in a more proactive manner in secondary prevention. Thereby, a more optimal state of functional capacity could be achieved, preventing or at least mitigating the risk for relapses. C: in the best-case scenario, programs aimed at healthy individuals in primary prevention would improve functional capacity to an optimal range before the onset of an incident, thereby conferring resilience to avoid or reduce pathological events and health care system dependence. Figure created with BioRender.com.


Financial relief, e.g., of health care costs, certainly is one of the strongest incentives. Moreover, the establishment of science-based, individualized, structured, and guided exercise programs, including aspects of behavior change and habit formation, would ensure the highest adherence and compliance (1262, 1263, 1269, 1281, 1282). This could be facilitated if general practitioners were able to issue physical activity or lifestyle prescriptions for inactive individuals, covered by health insurance. In most countries, this is usually only possible for individuals with existing physical impairments, thus in rehabilitation or secondary prevention rather than for primary prevention in those who are still healthy but inactive. The prevailing reactive health care system, focused on treatment and rehabilitation, leads to high costs and patient relapse (FIGURE 11A). Programs for secondary prevention that go beyond rehabilitation often do not exist but would help to minimize relapse by aiming at achieving functional capacity levels that surpass those that initially contributed to an incident or disease in these patients (FIGURE 11B). Optimally, proactive programs would be established for primary prevention in healthy individuals to reach a level of functional capacity and resilience that minimizes such incidents before they even occur (FIGURE 11C).

8.2.3. Health care professionals.

It is clear that a general recommendation of “be more active and eat a balanced diet” is insufficient (1283). Therefore, personalized physical activity or lifestyle counseling should be accessible for individuals with an unhealthy lifestyle, tailored for different populations such as geriatric individuals (1284, 1285). The recognition of the importance of physical activity is very divergent among health care professionals. For example, there is a large variation in exercise prescription by physicians depending on their own physical activity level (1283). In fact, physically active physicians provide almost twice as many daily physical activity consultations compared to their inactive colleagues. One of the major barriers for primary care physicians to prescribe physical activity to patients is a lack of education. Accordingly, medical curricula should much more strongly emphasize the importance of physical activity, which in many diseases is on par with pharmacological and other interventions (302). This should help health care professionals to incorporate routine fitness testing and exercise interventions in the clinic and guide patients toward structured programs (1286).

8.2.4. Individuals.

Education about the strong benefits of an active lifestyle has to be massively expanded, starting at young age, notably in an accessible and understandable manner to minimize misunderstanding and misinformation (1287). Often, such aspects are the first to be removed from already overloaded school curricula. Moreover, extracurricular activities, such as walk- or cycle-to-school programs, should also be strengthened, including the integration of road safety education (1271). Second, social safety nets, which reduce the work time of socioeconomically disadvantaged cohorts, would lessen time pressure and fatigue and increase motivation to engage in exercise (1288, 1289). The goal of the educational measures on different levels should be clear guidance for the design and application of personalized, evidence-based, safe, and efficacious training paradigms (1290, 1291), nutrition (1292), and other interventions.

8.3. Who Is Going to Pay for This?

Obviously, any of these measures is associated with considerable financial investments, at least in the short term, which might dampen the enthusiasm for such political and societal initiatives, e.g., in politicians who think in 2-, 4-, or 6-yr election cycles. It is important, however, to point out that in the long run better public health will yield enormous savings, even in the context of an aging population (233). For example, various studies estimate an average saving of USD 3–4 for each dollar invested in measures that promote physical activity alone (1293, 1294). Each increase in one metabolic equivalent (MET) in cardiorespiratory function leads to an individual annual reduction in health care costs between USD 1,025 and USD 5,193 in different populations (thus ∼5–10% of total costs) (1295, 1296), a decrease in all-cause sickness absence days (1297), improved work ability, and fewer doctor and hospital visits (1119). Further return of investment can be expected from other changes, e.g., healthy diet (1298), for example by lowering the annual costs of GBP 2.68 billion caused by unhealthy food in the United Kingdom (1299), cycling infrastructure (1300), or adequate sleep (1301). In Canada, the economic burden of low cardiorespiratory fitness is estimated at CAD 3.6 billion, with savings of CAD 644 million per year with a 10% reduction in the prevalence of low cardiorespiratory fitness (1302) and similar numbers for low muscle strength (CAD 3 billion total costs and CAD 546 million savings with 10% improvement) (1303). Along these lines, a reduction in incidents of fatal and nonfatal falls would save costs in the millions, if not billions, of dollars even only within the United States (1304). Finally, risk factors such as elevated BMI or waist circumference significantly drive health care costs, for example, by 15.4% more with an increase in waist circumference by 10 cm (1305). Thus, although finding a true “antiaging” drug or intervention could mean a lot of money to be gained (for a handful of individuals or companies), engaging in proven lifestyle- and behavior-associated interventions with certainty leads to a lot of money saved (for a society).

9. CONCLUSIONS AND OUTLOOK

All of these arguments should not be taken as a vote against current research into molecular clocks, epigenetic reprogramming and rejuvenation, and other cutting-edge topics in aging research. Hopefully, future insights in these fields will synergize with those obtained in research areas focused on the basic biology of the physiological biomarkers and together provide mechanistic and causal data on healthy and unhealthy aging. Moreover, potential age-reversal-age-extension (ARAE) paradoxical effects between pharmacological geroprotectors and lifestyle interventions, e.g., the attenuating effect of metformin, resveratrol, or rapamycin on training adaptation (37, 38, 475, 1306), could be overcome with a better understanding of the respective systems. Importantly, assessment and leverage of the physiological biomarkers (1307), the knowledge of proven drivers of unhealthy aging and of interventions promoting healthy aging are available now and could (should!) be applied immediately in an effort that would benefit all to attain a healthier, longer, and happier life (1308) (“a high tide lifts all boats”) instead of waiting for potential future breakthroughs, many of which might only be accessible to a handful of millionaires. Indeed, a three-round Delphi study recently came to the overwhelming conclusion that the physiological biomarkers are currently by far the best tools to monitor and assess interventional studies aimed at aging and longevity (1309). Until other biomarkers and interventions, e.g., those based on molecular criteria, reach the same level of maturity, it thus is important that, despite the heightened interest and the massive influx of money for basic research, biotechnological, and clinical application with the hope of immense return of investment (1310), hype of premature, preliminary, and not yet reproduced results should be avoided to temper inflated expectations in scientists, funders, the media, and the lay public alike (1311).

GRANTS

Our research group is supported by the Swiss National Science Foundation (CRSII5_209252), the Biozentrum, and the University of Basel.

DISCLOSURES

C.H. is an associate editor of Physiological Reviews and was not involved in and did not have access to information regarding the peer-review process or final disposition of this article. An alternate editor oversaw the peer-review and decision-making process for this article. None of the authors has any other conflicts of interest, financial or otherwise, to disclose.

AUTHOR CONTRIBUTIONS

R.F. and C.H. prepared figures; drafted manuscript; edited and revised manuscript; and approved final version of manuscript.

ACKNOWLEDGMENTS

We thank our colleagues from our research group for constructive feedback and input.

The figures were created with BioRender.com (all figures and graphical abstract) and Adobe Stock Photos (FIGURE 10 and graphical abstract).

REFERENCES

  • 1. Jeune B. Living longer—but better? Aging Clin Exp Res 14: 72–93, 2002. doi:10.1007/BF03324421.
    Crossref | PubMed | Web of Science | Google Scholar
  • 2. Gurven M, Kaplan H. Longevity among hunter-gatherers: a cross-cultural examination. Popul Dev Rev 33: 321–365, 2007. doi:10.1111/j.1728-4457.2007.00171.x.
    Crossref | Web of Science | Google Scholar
  • 3. Vaupel JW. Biodemography of human ageing. Nature 464: 536–542, 2010. doi:10.1038/nature08984.
    Crossref | PubMed | Web of Science | Google Scholar
  • 4. Dong X, Milholland B, Vijg J. Evidence for a limit to human lifespan. Nature 538: 257–259, 2016. doi:10.1038/nature19793.
    Crossref | PubMed | Web of Science | Google Scholar
  • 5. Modig K, Andersson T, Vaupel J, Rau R, Ahlbom A. How long do centenarians survive? Life expectancy and maximum lifespan. J Intern Med 282: 156–163, 2017. doi:10.1111/joim.12627.
    Crossref | PubMed | Web of Science | Google Scholar
  • 6. Christensen K, Doblhammer G, Rau R, Vaupel JW. Ageing populations: the challenges ahead. Lancet 374: 1196–1208, 2009. doi:10.1016/S0140-6736(09)61460-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 7. Eisenstein M. Does the human lifespan have a limit? Nature 601: S2–S4, 2022. doi:10.1038/d41586-022-00070-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 8. Rozing MP, Kirkwood TB, Westendorp RG. Is there evidence for a limit to human lifespan? Nature 546: E11–E12, 2017. doi:10.1038/nature22788.
    Crossref | PubMed | Web of Science | Google Scholar
  • 9. Lenart A, Vaupel JW. Questionable evidence for a limit to human lifespan. Nature 546: E13–E14, 2017. doi:10.1038/nature22790.
    Crossref | PubMed | Web of Science | Google Scholar
  • 10. Barbi E, Lagona F, Marsili M, Vaupel JW, Wachter KW. The plateau of human mortality: demography of longevity pioneers. Science 360: 1459–1461, 2018. doi:10.1126/science.aat3119.
    Crossref | PubMed | Web of Science | Google Scholar
  • 11. Olshansky SJ, Carnes BA. Inconvenient truths about human longevity. J Gerontol A Biol Sci Med Sci 74: S7–S12, 2019. doi:10.1093/gerona/glz098.
    Crossref | PubMed | Web of Science | Google Scholar
  • 12. Gbari S, Poulain M, Dal L, Denuit M. Extreme value analysis of mortality at the oldest ages: a case study based on individual ages at death. N Am Actuar J 21: 397–416, 2017. doi:10.1080/10920277.2017.1301260.
    Crossref | Google Scholar
  • 13. Einmahl JJ, Einmahl JH de Haan L. Limits to human life span through extreme value theory. J Am Stat Assoc 114: 1075–1080, 2019. doi:10.1080/01621459.2018.1537912.
    Crossref | Web of Science | Google Scholar
  • 14. Levine HJ. Rest heart rate and life expectancy. J Am Coll Cardiol 30: 1104–1106, 1997. doi:10.1016/s0735-1097(97)00246-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 15. Yun MH. Salamander insights into ageing and rejuvenation. Front Cell Dev Biol 9: 689062, 2021. doi:10.3389/fcell.2021.689062.
    Crossref | PubMed | Web of Science | Google Scholar
  • 16. Szekely P, Korem Y, Moran U, Mayo A, Alon U. The mass-longevity triangle: pareto optimality and the geometry of life-history trait space. PLoS Comput Biol 11: e1004524, 2015. doi:10.1371/journal.pcbi.1004524.
    Crossref | PubMed | Web of Science | Google Scholar
  • 17. Herculano-Houzel S. Longevity and sexual maturity vary across species with number of cortical neurons, and humans are no exception. J Comp Neurol 527: 1689–1705, 2019. doi:10.1002/cne.24564.
    Crossref | PubMed | Web of Science | Google Scholar
  • 18. Yuan R, Hascup E, Hascup K, Bartke A. Relationships among development, growth, body size, reproduction, aging, and longevity—trade-offs and pace-of-life. Biochemistry (Mosc) 88: 1692–1703, 2023. doi:10.1134/S0006297923110020.
    Crossref | PubMed | Web of Science | Google Scholar
  • 19. Crofts SJ, Latorre-Crespo E, Chandra T. DNA methylation rates scale with maximum lifespan across mammals. Nat Aging 4: 27–32, 2024. doi:10.1038/s43587-023-00535-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 20. Li CZ, Haghani A, Yan Q, Lu AT, Zhang J, Fei Z, Ernst J, Yang XW, Gladyshev VN, Robeck TR, Chavez AS, Cook JA, Dunnum JL, Raj K, Seluanov A, Gorbunova V, Horvath S. Epigenetic predictors of species maximum life span and other life-history traits in mammals. Sci Adv 10: eadm7273, 2024. doi:10.1126/sciadv.adm7273.
    Crossref | PubMed | Web of Science | Google Scholar
  • 21. Marck A, Antero J, Berthelot G, Saulière G, Jancovici JM, Masson-Delmotte V, Boeuf G, Spedding M, Le Bourg E, Toussaint JF. Are we reaching the limits of Homo sapiens? Front Physiol 8: 812, 2017. doi:10.3389/fphys.2017.00812.
    Crossref | PubMed | Web of Science | Google Scholar
  • 22. Adam D. Life expectancy rise in rich countries slows down: why discovery took 30 years to prove. Nature, 2024. doi:10.1038/d41586-024-03244-1.
    Crossref | PubMed | Google Scholar
  • 23. Milholland B. Jeanne Calment, actuarial paradoxography and the limit to human lifespan. Rejuvenation Res 23: 17–18, 2020. doi:10.1089/rej.2019.2272.
    Crossref | PubMed | Web of Science | Google Scholar
  • 24. Olshansky SJ, Willcox BJ, Demetrius L, Beltrán-Sánchez H. Implausibility of radical life extension in humans in the twenty-first century. Nat Aging 4: 1635–1642, 2024. doi:10.1038/s43587-024-00702-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 25. Beard JR, Hanewald K, Si Y, Amuthavalli Thiyagarajan J, Moreno-Agostino D. Cohort trends in intrinsic capacity in England and China. Nat Aging 5: 87–98, 2025. doi:10.1038/s43587-024-00741-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 26. Editors. Seventy really may be the new sixty for English baby boomers. Nat Aging 5: 19–20, 2025. doi:10.1038/s43587-024-00796-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 27. Garmany A, Terzic A. Global healthspan-lifespan gaps among 183 World Health Organization member states. JAMA Netw Open 7: e2450241, 2024. doi:10.1001/jamanetworkopen.2024.50241.
    Crossref | PubMed | Web of Science | Google Scholar
  • 28. Webster AJ, Clarke R. Sporadic, late-onset, and multistage diseases. PNAS Nexus 1: pgac095, 2022. doi:10.1093/pnasnexus/pgac095.
    Crossref | PubMed | Web of Science | Google Scholar
  • 29. Jones CH, Dolsten M. Healthcare on the brink: navigating the challenges of an aging society in the United States. NPJ Aging 10: 22, 2024. doi:10.1038/s41514-024-00148-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 30. Komp-Leukkunen K, Sarasma J. Social sustainability in aging populations: a systematic literature review. Gerontologist 64: gnad097, 2024. doi:10.1093/geront/gnad097.
    Crossref | PubMed | Web of Science | Google Scholar
  • 31. Jarzebski MP, Elmqvist T, Gasparatos A, Fukushi K, Eckersten S, Haase D, Goodness J, Khoshkar S, Saito O, Takeuchi K, Theorell T, Dong N, Kasuga F, Watanabe R, Sioen GB, Yokohari M, Pu J. Ageing and population shrinking: implications for sustainability in the urban century. NPJ Urban Sustain 1: 17, 2021. doi:10.1038/s42949-021-00023-z.
    Crossref | Web of Science | Google Scholar
  • 32. WHO. Ageing and health. World Health Organization (WHO), 2025. https://www.who.int/news-room/fact-sheets/detail/ageing-and-health [2025 Jan 16].
    Google Scholar
  • 33. Bloom DE, Zucker LM. Aging is the real population bomb. International Monetary Fund (IMF), 2025. https://www.imf.org/en/Publications/fandd/issues/Series/Analytical-Series/aging-is-the-real-population-bomb-bloom-zucker [2025 Jan 16].
    Google Scholar
  • 34. Poganik JR, Gladyshev VN. We need to shift the focus of aging research to aging itself. Proc Natl Acad Sci USA 120: e2307449120, 2023. doi:10.1073/pnas.2307449120.
    Crossref | PubMed | Web of Science | Google Scholar
  • 35. Meyer DH, Schumacher B. Aging clocks based on accumulating stochastic variation. Nat Aging 4: 871–885, 2024. doi:10.1038/s43587-024-00619-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 36. Ikram MA. The use and misuse of ‘biological aging’ in health research. Nat Med 30: 3045, 2024. doi:10.1038/s41591-024-03297-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 37. Furrer R, Handschin C. Lifestyle vs. pharmacological interventions for healthy aging. Aging (Albany NY) 12: 5–7, 2020. doi:10.18632/aging.102741.
    Crossref | PubMed | Web of Science | Google Scholar
  • 38. Furrer R, Handschin C. Drugs, clocks and exercise in ageing: hype and hope, fact and fiction. J Physiol 601: 2057–2068, 2023. doi:10.1113/JP282887.
    Crossref | PubMed | Web of Science | Google Scholar
  • 39. Johnson AA, Shokhirev MN. Contextualizing aging clocks and properly describing biological age. Aging Cell 23: e14377, 2024. doi:10.1111/acel.14377.
    Crossref | PubMed | Web of Science | Google Scholar
  • 40. Gladyshev VN, Anderson B, Barlit H, Barré B, Beck S, Behrouz B, , et al.. Disagreement on foundational principles of biological aging. PNAS Nexus 3: pgae499, 2024. doi:10.1093/pnasnexus/pgae499.
    Crossref | PubMed | Web of Science | Google Scholar
  • 41. Kaeberlein M. How healthy is the healthspan concept? Geroscience 40: 361–364, 2018. doi:10.1007/s11357-018-0036-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 42. Schramme T. Health as complete well-being: the WHO definition and beyond. Public Health Ethics 16: 210–218, 2023. doi:10.1093/phe/phad017.
    Crossref | PubMed | Web of Science | Google Scholar
  • 43. UN General Assembly. Decade of Healthy Aging: The Platform. United Nations General Assembly, 2025. https://www.decadeofhealthyageing.org/ [2025 Jan 3].
    Google Scholar
  • 44. World Health Organization. WHO’s work on the UN Decade of Healthy Ageing (2021–2030). World Health Organization, 2025. https://www.who.int/initiatives/decade-of-healthy-ageing [2025 Jan 3].
    Google Scholar
  • 45. Pamplona R, Jové M, Gómez J, Barja G. Programmed versus non-programmed evolution of aging. What is the evidence? Exp Gerontol 175: 112162, 2023. doi:10.1016/j.exger.2023.112162.
    Crossref | PubMed | Web of Science | Google Scholar
  • 46. Hawkes K. The centrality of ancestral grandmothering in human evolution. Integr Comp Biol 60: 765–781, 2020. doi:10.1093/icb/icaa029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 47. Hawkes K. Human longevity: the grandmother effect. Nature 428: 128–129, 2004. doi:10.1038/428128a.
    Crossref | PubMed | Web of Science | Google Scholar
  • 48. Lahdenperä M, Mar KU, Lummaa V. Reproductive cessation and post-reproductive lifespan in Asian elephants and pre-industrial humans. Front Zool 11: 54, 2014. doi:10.1186/s12983-014-0054-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 49. Ellis S, Franks DW, Nielsen ML, Weiss MN, Croft DP. The evolution of menopause in toothed whales. Nature 627: 579–585, 2024. doi:10.1038/s41586-024-07159-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 50. Wood BM, Negrey JD, Brown JL, Deschner T, Thompson ME, Gunter S, Mitani JC, Watts DP, Langergraber KE. Demographic and hormonal evidence for menopause in wild chimpanzees. Science 382: eadd5473, 2023. doi:10.1126/science.add5473.
    Crossref | PubMed | Web of Science | Google Scholar
  • 51. Winkler I, Goncalves A. Do mammals have menopause? Cell 186: 4729–4733, 2023. doi:10.1016/j.cell.2023.09.026.
    Crossref | PubMed | Web of Science | Google Scholar
  • 52. Vaill M, Kawanishi K, Varki N, Gagneux P, Varki A. Comparative physiological anthropogeny: exploring molecular underpinnings of distinctly human phenotypes. Physiol Rev 103: 2171–2229, 2023. doi:10.1152/physrev.00040.2021.
    Link | Web of Science | Google Scholar
  • 53. Coles LS. Demographics of human supercentenarians and the implications for longevity medicine. Ann NY Acad Sci 1019: 490–495, 2004. doi:10.1196/annals.1297.090.
    Crossref | PubMed | Web of Science | Google Scholar
  • 54. Perls T. Centenarian statistics, 2025. https://www.bumc.bu.edu/centenarian/statistics/ [2025 Jan 10].
    Google Scholar
  • 55. Melzer D, Pilling LC, Ferrucci L. The genetics of human ageing. Nat Rev Genet 21: 88–101, 2020. doi:10.1038/s41576-019-0183-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 56. Donlon TA, Morris BJ, Masaki KH, Chen R, Davy PM, Kallianpur KJ, Nakagawa K, Owens JB, Willcox DC, Allsopp RC, Willcox BJ. FOXO3, a resilience gene: impact on lifespan, healthspan, and deathspan. J Gerontol A Biol Sci Med Sci 77: 1479–1484, 2022. doi:10.1093/gerona/glac132.
    Crossref | PubMed | Web of Science | Google Scholar
  • 57. Zhang Y, Murata S, Schmidt-Mende K, Ebeling M, Modig K. Do people reach 100 by surviving, delaying, or avoiding diseases? A life course comparison of centenarians and non-centenarians from the same birth cohorts. Geroscience, 2024. doi:10.1007/s11357-024-01330-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 58. Marston HR, Niles-Yokum K, Silva PA. A commentary on Blue Zones®: a critical review of age-friendly environments in the 21st century and beyond. Int J Environ Res Public Health 18: 837, 2021. doi:10.3390/ijerph18020837.
    Crossref | PubMed | Web of Science | Google Scholar
  • 59. Poulain M, Pes GM, Grasland C, Carru C, Ferrucci L, Baggio G, Franceschi C, Deiana L. Identification of a geographic area characterized by extreme longevity in the Sardinia island: the AKEA study. Exp Gerontol 39: 1423–1429, 2004. doi:10.1016/j.exger.2004.06.016.
    Crossref | PubMed | Web of Science | Google Scholar
  • 60. Chiang CW, Marcus JH, Sidore C, Biddanda A, Al-Asadi H, Zoledziewska M, Pitzalis M, Busonero F, Maschio A, Pistis G, Steri M, Angius A, Lohmueller KE, Abecasis GR, Schlessinger D, Cucca F, Novembre J. Genomic history of the Sardinian population. Nat Genet 50: 1426–1434, 2018. doi:10.1038/s41588-018-0215-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 61. Pistis G, Piras I, Pirastu N, Persico I, Sassu A, Picciau A, Prodi D, Fraumene C, Mocci E, Manias MT, Atzeni R, Cosso M, Pirastu M, Angius A. High differentiation among eight villages in a secluded area of Sardinia revealed by genome-wide high density SNPs analysis. PLoS One 4: e4654, 2009. doi:10.1371/journal.pone.0004654.
    Crossref | PubMed | Web of Science | Google Scholar
  • 62. Ungvari Z, Fazekas-Pongor V, Csiszar A, Kunutsor SK. The multifaceted benefits of walking for healthy aging: from Blue Zones to molecular mechanisms. Geroscience 45: 3211–3239, 2023. doi:10.1007/s11357-023-00873-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 63. Pes GM, Dore MP, Tsofliou F, Poulain M. Diet and longevity in the Blue Zones: a set-and-forget issue? Maturitas 164: 31–37, 2022. doi:10.1016/j.maturitas.2022.06.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 64. Rosero-Bixby L. The vanishing advantage of longevity in Nicoya, Costa Rica: a cohort shift. Demogr Res 49: 723–736, 2023. doi:10.4054/DemRes.2023.49.27.
    Crossref | Web of Science | Google Scholar
  • 65. Hokama T, Binns C. Declining longevity advantage and low birthweight in Okinawa. Asia Pac J Public Health 20, Suppl: 95–101, 2008.
    PubMed | Google Scholar
  • 66. Le Bourg E. Dietary restriction studies in humans: focusing on obesity, forgetting longevity. Gerontology 58: 126–128, 2012. doi:10.1159/000328675.
    Crossref | PubMed | Web of Science | Google Scholar
  • 67. Gavrilova NS, Gavrilov LA. Comments on dietary restriction, Okinawa diet and longevity. Gerontology 58: 221–223, 2012. doi:10.1159/000329894.
    Crossref | PubMed | Web of Science | Google Scholar
  • 68. Poulain M. Exceptional longevity in Okinawa: a plea for in-depth validation. Demogr Res 25: 245–284, 2011. doi:10.4054/DemRes.2011.25.7.
    Crossref | Web of Science | Google Scholar
  • 69. Newman SJ. Supercentenarian and remarkable age records exhibit patterns indicative of clerical errors and pension fraud (Preprint). bioRxiv 704080, 2024. doi:10.1101/704080.
    Google Scholar
  • 70. Young RD, Desjardins B, McLaughlin K, Poulain M, Perls TT. Typologies of extreme longevity myths. Curr Gerontol Geriatr Res 2010: 423087, 2010. doi:10.1155/2010/423087.
    Crossref | PubMed | Google Scholar
  • 71. Newman SJ. The global pattern of centenarians highlights deep problems in demography (Preprint). medRxiv 2024.09.06.24313170, 2024. doi:10.1101/2024.09.06.24313170.
    Google Scholar
  • 72. Amigo I. Shades of blue. Science 386: 840–845, 2024. doi:10.1126/science.adu7169.
    Crossref | PubMed | Web of Science | Google Scholar
  • 73. Coppede F. Mutations involved in premature-ageing syndromes. Appl Clin Genet 14: 279–295, 2021. doi:10.2147/TACG.S273525.
    Crossref | PubMed | Web of Science | Google Scholar
  • 74. Austad SN. Methusaleh’s zoo: how nature provides us with clues for extending human health span. J Comp Pathol 142, Suppl 1: S10–S21, 2010. doi:10.1016/j.jcpa.2009.10.024.
    Crossref | PubMed | Web of Science | Google Scholar
  • 75. Herskind AM, McGue M, Holm NV, Sørensen TI, Harvald B, Vaupel JW. The heritability of human longevity: a population-based study of 2872 Danish twin pairs born 1870-1900. Hum Genet 97: 319–323, 1996. doi:10.1007/BF02185763.
    Crossref | PubMed | Web of Science | Google Scholar
  • 76. Ruby JG, Wright KM, Rand KA, Kermany A, Noto K, Curtis D, Varner N, Garrigan D, Slinkov D, Dorfman I, Granka JM, Byrnes J, Myres N, Ball C. Estimates of the heritability of human longevity are substantially inflated due to assortative mating. Genetics 210: 1109–1124, 2018. doi:10.1534/genetics.118.301613.
    Crossref | PubMed | Web of Science | Google Scholar
  • 77. Gubbi S, Schwartz E, Crandall J, Verghese J, Holtzer R, Atzmon G, Braunstein R, Barzilai N, Milman S. Effect of exceptional parental longevity and lifestyle factors on prevalence of cardiovascular disease in offspring. Am J Cardiol 120: 2170–2175, 2017. doi:10.1016/j.amjcard.2017.08.040.
    Crossref | PubMed | Web of Science | Google Scholar
  • 78. Rajpathak SN, Liu Y, Ben-David O, Reddy S, Atzmon G, Crandall J, Barzilai N. Lifestyle factors of people with exceptional longevity. J Am Geriatr Soc 59: 1509–1512, 2011. doi:10.1111/j.1532-5415.2011.03498.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 79. Wojczynski MK, Jiuan Lin S, Sebastiani P, Perls TT, Lee J, Kulminski A, Newman A, Zmuda JM, Christensen K, Province MA. NIA Long Life Family Study: objectives, design, and heritability of cross-sectional and longitudinal phenotypes. J Gerontol A Biol Sci Med Sci 77: 717–727, 2022. doi:10.1093/gerona/glab333.
    Crossref | PubMed | Web of Science | Google Scholar
  • 80. Toniolo L, Sirago G, Giacomello E. Experimental models for ageing research. Histol Histopathol 38: 597–605, 2023. doi:10.14670/HH-18-576.
    Crossref | PubMed | Web of Science | Google Scholar
  • 81. Mitchell SJ, Scheibye-Knudsen M, Longo DL, de Cabo R. Animal models of aging research: implications for human aging and age-related diseases. Annu Rev Anim Biosci 3: 283–303, 2015. doi:10.1146/annurev-animal-022114-110829.
    Crossref | PubMed | Web of Science | Google Scholar
  • 82. Austad SN, Podlutsky A. A critical evaluation of nonmammalian models for aging research. In: Handbook of the Biology of Aging, edited by Masoro EJ, Austad SN. Academic Press, 2006, p. 449–467. doi:10.1016/B978-0-12-088387-5.X5000-6.
    Crossref | Google Scholar
  • 83. Wolf AM. Rodent diet aids and the fallacy of caloric restriction. Mech Ageing Dev 200: 111584, 2021. doi:10.1016/j.mad.2021.111584.
    Crossref | PubMed | Web of Science | Google Scholar
  • 84. Martin B, Ji S, Maudsley S, Mattson MP. “Control” laboratory rodents are metabolically morbid: why it matters. Proc Natl Acad Sci USA 107: 6127–6133, 2010. doi:10.1073/pnas.0912955107.
    Crossref | PubMed | Web of Science | Google Scholar
  • 85. Panowski SH, Dillin A. Signals of youth: endocrine regulation of aging in Caenorhabditis elegans. Trends Endocrinol Metab 20: 259–264, 2009. doi:10.1016/j.tem.2009.03.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 86. Kern CC, Srivastava S, Ezcurra M, Hsiung KC, Hui N, Townsend S, Maczik D, Zhang B, Tse V, Konstantellos V, Bähler J, Gems D. C. elegans ageing is accelerated by a self-destructive reproductive programme. Nat Commun 14: 4381, 2023. doi:10.1038/s41467-023-40088-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 87. Perlman RL. Mouse models of human disease: an evolutionary perspective. Evol Med Public Health 2016: 170–176, 2016. doi:10.1093/emph/eow014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 88. Agoston DV. How to translate time? The temporal aspect of human and rodent biology. Front Neurol 8: 92, 2017. doi:10.3389/fneur.2017.00092.
    Crossref | PubMed | Web of Science | Google Scholar
  • 89. Oeseburg H, de Boer RA, van Gilst WH, van der Harst P. Telomere biology in healthy aging and disease. Pflugers Arch 459: 259–268, 2010. doi:10.1007/s00424-009-0728-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 90. Cohen AA. Aging across the tree of life: the importance of a comparative perspective for the use of animal models in aging. Biochim Biophys Acta Mol Basis Dis 1864: 2680–2689, 2018. doi:10.1016/j.bbadis.2017.05.028.
    Crossref | PubMed | Web of Science | Google Scholar
  • 91. Edwards JE, Hiltz E, Broell F, Bushnell PG, Campana SE, Christiansen JS, Devine BM, Gallant JJ, Hedges KJ, MacNeil MA, McMeans BC, Nielsen J, Præbel K, Skomal GB, Steffensen JF, Walter RP, Watanabe YY, VanderZwaag DL, Hussey NE. Advancing research for the management of long-lived species: a case study on the Greenland shark. Front Mar Sci 6, 2019. doi:10.3389/fmars.2019.00087.
    Crossref | Web of Science | Google Scholar
  • 92. Holtze S, Gorshkova E, Braude S, Cellerino A, Dammann P, Hildebrandt TB, Hoeflich A, Hoffmann S, Koch P, Terzibasi Tozzini E, Skulachev M, Skulachev VP, Sahm A. Alternative animal models of aging research. Front Mol Biosci 8: 660959, 2021. doi:10.3389/fmolb.2021.660959.
    Crossref | PubMed | Web of Science | Google Scholar
  • 93. Jové M, Mota-Martorell N, Fernández-Bernal A, Portero-Otin M, Barja G, Pamplona R. Phenotypic molecular features of long-lived animal species. Free Radic Biol Med 208: 728–747, 2023. doi:10.1016/j.freeradbiomed.2023.09.023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 94. Lee MB, Hill CM, Bitto A, Kaeberlein M. Antiaging diets: separating fact from fiction. Science 374: eabe7365, 2021. doi:10.1126/science.abe7365.
    Crossref | PubMed | Web of Science | Google Scholar
  • 95. Ganeshan K, Chawla A. Warming the mouse to model human diseases. Nat Rev Endocrinol 13: 458–465, 2017. doi:10.1038/nrendo.2017.48.
    Crossref | PubMed | Web of Science | Google Scholar
  • 96. Škop V, Guo J, Liu N, Xiao C, Hall KD, Gavrilova O, Reitman ML. Mouse thermoregulation: introducing the concept of the thermoneutral point. Cell Rep 31: 107501, 2020. doi:10.1016/j.celrep.2020.03.065.
    Crossref | PubMed | Web of Science | Google Scholar
  • 97. Booth FW, Laye MJ. Lack of adequate appreciation of physical exercise's complexities can pre-empt appropriate design and interpretation in scientific discovery. J Physiol 587: 5527–5539, 2009. doi:10.1113/jphysiol.2009.179507.
    Crossref | PubMed | Web of Science | Google Scholar
  • 98. Widjaja AA, Lim WW, Viswanathan S, Chothani S, Corden B, Dasan CM, Goh JW, Lim R, Singh BK, Tan J, Pua CJ, Lim SY, Adami E, Schafer S, George BL, Sweeney M, Xie C, Tripathi M, Sims NA, Hübner N, Petretto E, Withers DJ, Ho L, Gil J, Carling D, Cook SA. Inhibition of IL-11 signalling extends mammalian healthspan and lifespan. Nature 632: 157–165, 2024. doi:10.1038/s41586-024-07701-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 99. Miller RA. Blocking an inflammatory protein slows the pace of ageing. Nature 632: 35–36, 2024. doi:10.1038/d41586-024-02300-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 100. Metcalfe RD, Putoczki TL, Griffin MD. Structural understanding of interleukin 6 family cytokine signaling and targeted therapies: focus on interleukin 11. Front Immunol 11: 1424, 2020. doi:10.3389/fimmu.2020.01424.
    Crossref | PubMed | Web of Science | Google Scholar
  • 101. Langston PK, Mathis D. Immunological regulation of skeletal muscle adaptation to exercise. Cell Metab 36: 1175–1183, 2024. doi:10.1016/j.cmet.2024.04.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 102. Alic N, Partridge L. Antagonizing Methuselah to extend life span. Genome Biol 8: 222, 2007. doi:10.1186/gb-2007-8-8-222.
    Crossref | PubMed | Web of Science | Google Scholar
  • 103. Gems D, Partridge L. Genetics of longevity in model organisms: debates and paradigm shifts. Annu Rev Physiol 75: 621–644, 2013. doi:10.1146/annurev-physiol-030212-183712.
    Crossref | PubMed | Web of Science | Google Scholar
  • 104. Valencak TG, Spenlingwimmer T, Nimphy R, Reinisch I, Hoffman JM, Prokesch A. Challenging a “cushy” life: potential roles of thermogenesis and adipose tissue adaptations in delayed aging of Ames and Snell Dwarf mice. Metabolites 10: 176, 2020. doi:10.3390/metabo10050176.
    Crossref | PubMed | Web of Science | Google Scholar
  • 105. Flurkey K, Papaconstantinou J, Harrison DE. The Snell dwarf mutation Pit1(dw) can increase life span in mice. Mech Ageing Dev 123: 121–130, 2002. doi:10.1016/s0047-6374(01)00339-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 106. Bartke A. Growth hormone and aging. Rev Endocr Metab Disord 22: 71–80, 2021. doi:10.1007/s11154-020-09593-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 107. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, Wei M, Madia F, Cheng CW, Hwang D, Martin-Montalvo A, Saavedra J, Ingles S, de Cabo R, Cohen P, Longo VD. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med 3: 70ra13, 2011. doi:10.1126/scitranslmed.3001845.
    Crossref | PubMed | Web of Science | Google Scholar
  • 108. Bartke A, Quainoo N. Impact of growth hormone-related mutations on mammalian aging. Front Genet 9: 586, 2018. doi:10.3389/fgene.2018.00586.
    Crossref | PubMed | Web of Science | Google Scholar
  • 109. Aguiar-Oliveira MH, Bartke A. Growth hormone deficiency: health and longevity. Endocr Rev 40: 575–601, 2019. doi:10.1210/er.2018-00216.
    Crossref | PubMed | Web of Science | Google Scholar
  • 110. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell 186: 243–278, 2023. doi:10.1016/j.cell.2022.11.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 111. Gems D, de Magalhães JP. The hoverfly and the wasp: A critique of the hallmarks of aging as a paradigm. Ageing Res Rev 70: 101407, 2021. doi:10.1016/j.arr.2021.101407.
    Crossref | PubMed | Web of Science | Google Scholar
  • 112. Bellantuono I. Find drugs that delay many diseases of old age. Nature 554: 293–295, 2018. doi:10.1038/d41586-018-01668-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 113. Keshavarz M, Xie K, Schaaf K, Bano D, Ehninger D. Targeting the “hallmarks of aging” to slow aging and treat age-related disease: fact or fiction? Mol Psychiatry 28: 242–255, 2023. doi:10.1038/s41380-022-01680-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 114. Mishra SK, Balendra V, Esposto J, Obaid AA, Maccioni RB, Jha NK, Perry G, Moustafa M, Al-Shehri M, Singh MP, Khan AA, Vamanu E, Singh SK. Therapeutic antiaging strategies. Biomedicines 10: 2515, 2022. doi:10.3390/biomedicines10102515.
    Crossref | PubMed | Web of Science | Google Scholar
  • 115. De Sousa Lages A, Lopes V, Horta J, Espregueira-Mendes J, Andrade R, Rebelo-Marques A. Therapeutics that can potentially replicate or augment the anti-aging effects of physical exercise. Int J Mol Sci 23: 9957, 2022. doi:10.3390/ijms23179957.
    Crossref | PubMed | Web of Science | Google Scholar
  • 116. Chaudhari PS, Ermolaeva MA. Too old for healthy aging? Exploring age limits of longevity treatments. NPJ Metab Health Dis 2: 37, 2024. doi:10.1038/s44324-024-00040-3.
    Crossref | PubMed | Google Scholar
  • 117. Strong R, Miller RA, Antebi A, Astle CM, Bogue M, Denzel MS, Fernandez E, Flurkey K, Hamilton KL, Lamming DW, Javors MA, de Magalhães JP, Martinez PA, McCord JM, Miller BF, Müller M, Nelson JF, Ndukum J, Rainger GE, Richardson A, Sabatini DM, Salmon AB, Simpkins JW, Steegenga WT, Nadon NL, Harrison DE. Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an alpha-glucosidase inhibitor or a Nrf2-inducer. Aging Cell 15: 872–884, 2016. doi:10.1111/acel.12496.
    Crossref | PubMed | Web of Science | Google Scholar
  • 118. Barinda AJ, Hardi H, Louisa M, Khatimah NG, Marliau RM, Felix I, Fadhillah MR, Jamal AK. Repurposing effect of cardiovascular-metabolic drug to increase lifespan: a systematic review of animal studies and current clinical trial progress. Front Pharmacol 15: 1373458, 2024. doi:10.3389/fphar.2024.1373458.
    Crossref | PubMed | Web of Science | Google Scholar
  • 119. Miller RA, Harrison DE, Allison DB, Bogue M, Debarba L, Diaz V, Fernandez E, Galecki A, Garvey WT, Jayarathne H, Kumar N, Javors MA, Ladiges WC, Macchiarini F, Nelson J, Reifsnyder P, Rosenthal NA, Sadagurski M, Salmon AB, Smith DL Jr, Snyder JM, Lombard DB, Strong R. Canagliflozin extends life span in genetically heterogeneous male but not female mice. JCI Insight 5: e140019, 2020. doi:10.1172/jci.insight.140019.
    Crossref | Web of Science | Google Scholar
  • 120. Harrison DE, Strong R, Reifsnyder P, Rosenthal N, Korstanje R, Fernandez E, Flurkey K, Ginsburg BC, Murrell MD, Javors MA, Lopez-Cruzan M, Nelson JF, Willcox BJ, Allsopp R, Watumull DM, Watumull DG, Cortopassi G, Kirkland JL, Tchkonia T, Choi YG, Yousefzadeh MJ, Robbins PD, Mitchell JR, Acar M, Sarnoski EA, Bene MR, Salmon A, Kumar N, Miller RA. Astaxanthin and meclizine extend lifespan in UM-HET3 male mice; fisetin, SG1002 (hydrogen sulfide donor), dimethyl fumarate, mycophenolic acid, and 4-phenylbutyrate do not significantly affect lifespan in either sex at the doses and schedules used. Geroscience 46: 795–816, 2024. doi:10.1007/s11357-023-01011-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 121. Bin P, Zhu C, Liu S, Li Z, Ren W, Zhu G. Perspective: methionine restriction-induced longevity-a possible role for inhibiting the synthesis of bacterial quorum sensing molecules. Adv Nutr 11: 773–783, 2020. doi:10.1093/advances/nmaa028.
    Crossref | PubMed | Web of Science | Google Scholar
  • 122. Dang Y, An Y, He J, Huang B, Zhu J, Gao M, Zhang S, Wang X, Yang B, Xie Z. Berberine ameliorates cellular senescence and extends the lifespan of mice via regulating p16 and cyclin protein expression. Aging Cell 19: e13060, 2020. doi:10.1111/acel.13060.
    Crossref | PubMed | Web of Science | Google Scholar
  • 123. Johnson AA, Cuellar TL. Glycine and aging: evidence and mechanisms. Ageing Res Rev 87: 101922, 2023. doi:10.1016/j.arr.2023.101922.
    Crossref | PubMed | Web of Science | Google Scholar
  • 124. Xue H, Thaivalappil A, Cao K. The potentials of methylene blue as an anti-aging drug. Cells 10: 3379, 2021. doi:10.3390/cells10123379.
    Crossref | PubMed | Web of Science | Google Scholar
  • 125. Mohammadinejad R, Ahmadi Z, Tavakol S, Ashrafizadeh M. Berberine as a potential autophagy modulator. J Cell Physiol 234: 14914–14926, 2019. doi:10.1002/jcp.28325.
    Crossref | PubMed | Web of Science | Google Scholar
  • 126. Mondal SA, Mann SN, van der Linden C, Sathiaseelan R, Kamal M, Das S, Bubak MP, Logan S, Miller BF, Stout MB. Metabolic benefits of 17alpha-estradiol in liver are partially mediated by ERbeta in male mice. Sci Rep 13: 9841, 2023. doi:10.1038/s41598-023-37007-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 127. La Grotta R, Frigé C, Matacchione G, Olivieri F, de Candia P, Ceriello A, Prattichizzo F. Repurposing SGLT-2 inhibitors to target aging: available evidence and molecular mechanisms. Int J Mol Sci 23: 12325, 2022. doi:10.3390/ijms232012325.
    Crossref | PubMed | Web of Science | Google Scholar
  • 128. Strong R, Miller RA, Astle CM, Floyd RA, Flurkey K, Hensley KL, Javors MA, Leeuwenburgh C, Nelson JF, Ongini E, Nadon NL, Warner HR, Harrison DE. Nordihydroguaiaretic acid and aspirin increase lifespan of genetically heterogeneous male mice. Aging Cell 7: 641–650, 2008. doi:10.1111/j.1474-9726.2008.00414.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 129. Knufinke M, MacArthur MR, Ewald CY, Mitchell SJ. Sex differences in pharmacological interventions and their effects on lifespan and healthspan outcomes: a systematic review. Front Aging 4: 1172789, 2023. doi:10.3389/fragi.2023.1172789.
    Crossref | PubMed | Web of Science | Google Scholar
  • 130. Bene M, Salmon AB. Testing the evidence that lifespan-extending compound interventions are conserved across laboratory animal model species. Geroscience 45: 1401–1409, 2023. doi:10.1007/s11357-022-00722-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 131. Lee MB, Blue B, Muir M, Kaeberlein M. The million-molecule challenge: a moonshot project to rapidly advance longevity intervention discovery. Geroscience 45: 3103–3113, 2023. doi:10.1007/s11357-023-00867-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 132. Sharp ZD, Strong R. Rapamycin, the only drug that has been consistently demonstrated to increase mammalian longevity. An update. Exp Gerontol 176: 112166, 2023. doi:10.1016/j.exger.2023.112166.
    Crossref | PubMed | Web of Science | Google Scholar
  • 133. Leslie M. Hunt for longevity drugs gets new life. Science 385: 1148–1149, 2024. doi:10.1126/science.adt0783.
    Crossref | PubMed | Web of Science | Google Scholar
  • 134. Leslie M. Hunt for longevity drugs gets new life. Science 385: 1148–1149, 2024. doi:10.1126/science.adt0783.
    Crossref | PubMed | Web of Science | Google Scholar
  • 135. Biomarkers of Aging Consortium, Herzog CM, Goeminne LJ, Poganik JR, Barzilai N, Belsky DW, , et al.. Challenges and recommendations for the translation of biomarkers of aging. Nat Aging 4: 1372–1383, 2024. doi:10.1038/s43587-024-00683-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 136. Finkel T. Ageing: a toast to long life. Nature 425: 132–133, 2003. doi:10.1038/425132a.
    Crossref | PubMed | Web of Science | Google Scholar
  • 137. Hall SS. Longevity research. In vino vitalis? Compounds activate life-extending genes. Science 301: 1165, 2003. doi:10.1126/science.301.5637.1165.
    Crossref | PubMed | Web of Science | Google Scholar
  • 138. Miller RA, Harrison DE, Astle CM, Baur JA, Boyd AR, de Cabo R, Fernandez E, Flurkey K, Javors MA, Nelson JF, Orihuela CJ, Pletcher S, Sharp ZD, Sinclair D, Starnes JW, Wilkinson JE, Nadon NL, Strong R. Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci 66: 191–201, 2011. doi:10.1093/gerona/glq178.
    Crossref | PubMed | Web of Science | Google Scholar
  • 139. Bass TM, Weinkove D, Houthoofd K, Gems D, Partridge L. Effects of resveratrol on lifespan in Drosophila melanogaster and Caenorhabditis elegans. Mech Ageing Dev 128: 546–552, 2007. doi:10.1016/j.mad.2007.07.007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 140. Kaeberlein M, McDonagh T, Heltweg B, Hixon J, Westman EA, Caldwell SD, Napper A, Curtis R, DiStefano PS, Fields S, Bedalov A, Kennedy BK. Substrate-specific activation of sirtuins by resveratrol. J Biol Chem 280: 17038–17045, 2005. doi:10.1074/jbc.M500655200.
    Crossref | PubMed | Web of Science | Google Scholar
  • 141. Beher D, Wu J, Cumine S, Kim KW, Lu SC, Atangan L, Wang M. Resveratrol is not a direct activator of SIRT1 enzyme activity. Chem Biol Drug Des 74: 619–624, 2009. doi:10.1111/j.1747-0285.2009.00901.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 142. Pacholec M, Bleasdale JE, Chrunyk B, Cunningham D, Flynn D, Garofalo RS, Griffith D, Griffor M, Loulakis P, Pabst B, Qiu X, Stockman B, Thanabal V, Varghese A, Ward J, Withka J, Ahn K. SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J Biol Chem 285: 8340–8351, 2010. doi:10.1074/jbc.M109.088682.
    Crossref | PubMed | Web of Science | Google Scholar
  • 143. Brenner C. Sirtuins are not conserved longevity genes. Life Metab 1: 122–133, 2022. doi:10.1093/lifemeta/loac025.
    Crossref | PubMed | Web of Science | Google Scholar
  • 144. Schmidt C. GSK/Sirtris compounds dogged by assay artifacts. Nat Biotechnol 28: 185–186, 2010. doi:10.1038/nbt0310-185.
    Crossref | PubMed | Web of Science | Google Scholar
  • 145. Shaito A, Posadino AM, Younes N, Hasan H, Halabi S, Alhababi D, Al-Mohannadi A, Abdel-Rahman WM, Eid AH, Nasrallah GK, Pintus G. Potential adverse effects of resveratrol: a literature review. Int J Mol Sci 21: 2084, 2020. doi:10.3390/ijms21062084.
    Crossref | PubMed | Web of Science | Google Scholar
  • 146. Pezzuto JM. Resveratrol: twenty years of growth, development and controversy. Biomol Ther (Seoul) 27: 1–14, 2019. doi:10.4062/biomolther.2018.176.
    Crossref | PubMed | Web of Science | Google Scholar
  • 147. Tomé-Carneiro J, Larrosa M, González-Sarriás A, Tomás-Barberán FA, García-Conesa MT, Espín JC. Resveratrol and clinical trials: the crossroad from in vitro studies to human evidence. Curr Pharm Des 19: 6064–6093, 2013. doi:10.2174/13816128113199990407.
    Crossref | PubMed | Web of Science | Google Scholar
  • 148. Cai T, Hu Y, Ding B, Yan R, Liu B, Cai L, Jing T, Jiang L, Xie X, Wang Y, Wang H, Zhou Y, He K, Xu L, Chen L, Cheng C, Ma J. Effect of metformin on testosterone levels in male patients with type 2 diabetes mellitus treated with insulin. Front Endocrinol (Lausanne) 12: 813067, 2021. doi:10.3389/fendo.2021.813067.
    Crossref | PubMed | Web of Science | Google Scholar
  • 149. Mohammed I, Hollenberg MD, Ding H, Triggle CR. A critical review of the evidence that metformin is a putative anti-aging drug that enhances healthspan and extends lifespan. Front Endocrinol (Lausanne) 12: 718942, 2021. doi:10.3389/fendo.2021.718942.
    Crossref | PubMed | Web of Science | Google Scholar
  • 150. Parish AJ, Swindell WR. Metformin has heterogeneous effects on model organism lifespans and is beneficial when started at an early age in Caenorhabditis elegans: a systematic review and meta-analysis. Aging Cell 21: e13733, 2022. doi:10.1111/acel.13733.
    Crossref | PubMed | Web of Science | Google Scholar
  • 151. Keys MT, Thinggaard M, Larsen LA, Pedersen DA, Hallas J, Christensen K. Reassessing the evidence of a survival advantage in type 2 diabetes treated with metformin compared with controls without diabetes: a retrospective cohort study. Int J Epidemiol 51: 1886–1898, 2022. doi:10.1093/ije/dyac200.
    Crossref | PubMed | Web of Science | Google Scholar
  • 152. Lee CG, Heckman-Stoddard B, Dabelea D, Gadde KM, Ehrmann D, Ford L, Prorok P, Boyko EJ, Pi-Sunyer X, Wallia A, Knowler WC, Crandall JP, Temprosa M; Diabetes Prevention Program Research Group and Diabetes Prevention Program Research Group. Effect of metformin and lifestyle interventions on mortality in the Diabetes Prevention Program and Diabetes Prevention Program Outcomes Study. Diabetes Care 44: 2775–2782, 2021. doi:10.2337/dc21-1046.
    Crossref | PubMed | Web of Science | Google Scholar
  • 153. Triggle CR, Mohammed I, Bshesh K, Marei I, Ye K, Ding H, MacDonald R, Hollenberg MD, Hill MA. Metformin: is it a drug for all reasons and diseases? Metabolism 133: 155223, 2022. doi:10.1016/j.metabol.2022.155223.
    Crossref | PubMed | Web of Science | Google Scholar
  • 154. Yang Y, Lu X, Liu N, Ma S, Zhang H, Zhang Z, , et al.. Metformin decelerates aging clock in male monkeys. Cell 187: 6358–6378.e29, 2024. doi:10.1016/j.cell.2024.08.021.
    Crossref | PubMed | Web of Science | Google Scholar
  • 155. Kozlov M. The brain aged more slowly in monkeys given a cheap diabetes drug. Nature, 2024. doi:10.1038/d41586-024-02938-w.
    Crossref | PubMed | Google Scholar
  • 156. Wang D, Eisen HJ. Mechanistic target of rapamycin (mTOR) inhibitors. Handb Exp Pharmacol 272: 53–72, 2022. doi:10.1007/164_2021_553.
    Crossref | PubMed | Google Scholar
  • 157. Kaeberlein M. Rapamycin and ageing: when, for how long, and how much? J Genet Genomics 41: 459–463, 2014. doi:10.1016/j.jgg.2014.06.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 158. Ham DJ, Börsch A, Lin S, Thürkauf M, Weihrauch M, Reinhard JR, Delezie J, Battilana F, Wang X, Kaiser MS, Guridi M, Sinnreich M, Rich MM, Mittal N, Tintignac LA, Handschin C, Zavolan M, Ruegg MA. The neuromuscular junction is a focal point of mTORC1 signaling in sarcopenia. Nat Commun 11: 4510, 2020. doi:10.1038/s41467-020-18140-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 159. Ham DJ, Börsch A, Chojnowska K, Lin S, Leuchtmann AB, Ham AS, Thürkauf M, Delezie J, Furrer R, Burri D, Sinnreich M, Handschin C, Tintignac LA, Zavolan M, Mittal N, Ruegg MA. Distinct and additive effects of calorie restriction and rapamycin in aging skeletal muscle. Nat Commun 13: 2025, 2022. doi:10.1038/s41467-022-29714-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 160. Unnikrishnan A, Kurup K, Salmon AB, Richardson A. Is rapamycin a dietary restriction mimetic? J Gerontol A Biol Sci Med Sci 75: 4–13, 2020. doi:10.1093/gerona/glz060.
    Crossref | PubMed | Web of Science | Google Scholar
  • 161. Orenduff MC, Coleman MF, Glenny EM, Huffman KM, Rezeli ET, Bareja A, Pieper CF, Kraus VB, Hursting SD. Differential effects of calorie restriction and rapamycin on age-related molecular and functional changes in skeletal muscle. Exp Gerontol 165: 111841, 2022. doi:10.1016/j.exger.2022.111841.
    Crossref | PubMed | Web of Science | Google Scholar
  • 162. Lee DJ, Hodzic Kuerec A, Maier AB. Targeting ageing with rapamycin and its derivatives in humans: a systematic review. Lancet Healthy Longev 5: e152–e162, 2024. doi:10.1016/S2666-7568(23)00258-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 163. Trelinska J, Dachowska I, Kotulska K, Fendler W, Jozwiak S, Mlynarski W. Complications of mammalian target of rapamycin inhibitor anticancer treatment among patients with tuberous sclerosis complex are common and occasionally life-threatening. Anticancer Drugs 26: 437–442, 2015. doi:10.1097/CAD.0000000000000207.
    Crossref | PubMed | Web of Science | Google Scholar
  • 164. Kaeberlein M. RTB101 and immune function in the elderly: interpreting an unsuccessful clinical trial. Transl Med Aging 4: 32–34, 2020. doi:10.1016/j.tma.2020.01.002.
    Crossref | Google Scholar
  • 165. Faes S, Demartines N, Dormond O. Mechanistic target of rapamycin inhibitors in renal cell carcinoma: potential, limitations, and perspectives. Front Cell Dev Biol 9: 636037, 2021. doi:10.3389/fcell.2021.636037.
    Crossref | PubMed | Web of Science | Google Scholar
  • 166. Grunewald M, Kumar S, Sharife H, Volinsky E, Gileles-Hillel A, Licht T, Permyakova A, Hinden L, Azar S, Friedmann Y, Kupetz P, Tzuberi R, Anisimov A, Alitalo K, Horwitz M, Leebhoff S, Khoma OZ, Hlushchuk R, Djonov V, Abramovitch R, Tam J, Keshet E. Counteracting age-related VEGF signaling insufficiency promotes healthy aging and extends life span. Science 373: eabc8479, 2021. doi:10.1126/science.abc8479.
    Crossref | PubMed | Web of Science | Google Scholar
  • 167. Paquette M, El-Houjeiri L, Pause A. mTOR pathways in cancer and autophagy. Cancers (Basel) 10, 2018. doi:10.3390/cancers10010018.
    Crossref | PubMed | Web of Science | Google Scholar
  • 168. Mabood Khalil MA, Al-Ghamdi SM, Dawood US, Ahmed Khamis SS, Ishida H, Chong VH, Tan J. Mammalian target of rapamycin inhibitors and wound healing complications in kidney transplantation: old myths and new realities. J Transplant 2022: 6255339, 2022. doi:10.1155/2022/6255339.
    Crossref | PubMed | Web of Science | Google Scholar
  • 169. Triana P, López-Gutierrez JC. Menstrual disorders associated with sirolimus treatment. Pediatr Blood Cancer 68: e28867, 2021. doi:10.1002/pbc.28867.
    Crossref | PubMed | Web of Science | Google Scholar
  • 170. Guo Z, Yu Q. Role of mTOR signaling in female reproduction. Front Endocrinol (Lausanne) 10: 692, 2019. doi:10.3389/fendo.2019.00692.
    Crossref | PubMed | Web of Science | Google Scholar
  • 171. Oliveira PF, Cheng CY, Alves MG. Emerging role for mammalian target of rapamycin in male fertility. Trends Endocrinol Metab 28: 165–167, 2017. doi:10.1016/j.tem.2016.12.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 172. Kaplan B, Qazi Y, Wellen JR. Strategies for the management of adverse events associated with mTOR inhibitors. Transplant Rev (Orlando) 28: 126–133, 2014. doi:10.1016/j.trre.2014.03.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 173. Johnston O, Rose CL, Webster AC, Gill JS. Sirolimus is associated with new-onset diabetes in kidney transplant recipients. J Am Soc Nephrol 19: 1411–1418, 2008. doi:10.1681/ASN.2007111202.
    Crossref | PubMed | Web of Science | Google Scholar
  • 174. Teutonico A, Schena PF, Di Paolo S. Glucose metabolism in renal transplant recipients: effect of calcineurin inhibitor withdrawal and conversion to sirolimus. J Am Soc Nephrol 16: 3128–3135, 2005. doi:10.1681/ASN.2005050487.
    Crossref | PubMed | Web of Science | Google Scholar
  • 175. Shi Q, Chang C, Saliba A, Bhat MA. Microglial mTOR activation upregulates Trem2 and Enhances beta-Amyloid Plaque clearance in the 5XFAD Alzheimer’s disease model. J Neurosci 42: 5294–5313, 2022. doi:10.1523/JNEUROSCI.2427-21.2022.
    Crossref | PubMed | Web of Science | Google Scholar
  • 176. Di Micco R, Krizhanovsky V, Baker D, d’Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol 22: 75–95, 2021. doi:10.1038/s41580-020-00314-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 177. Davan-Wetton CS, Pessolano E, Perretti M, Montero-Melendez T. Senescence under appraisal: hopes and challenges revisited. Cell Mol Life Sci 78: 3333–3354, 2021. doi:10.1007/s00018-020-03746-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 178. Zhao H, Liu Z, Chen H, Han M, Zhang M, Liu K, Jin H, Liu X, Shi M, Pu W, Werner M, Meister M, Kauschke SG, Sun R, Wang J, Shen R, Wang QD, Ma X, Tchorz JS, Zhou B. Identifying specific functional roles for senescence across cell types. Cell 187: 7314–7334.e21, 2024. doi:10.1016/j.cell.2024.09.021.
    Crossref | PubMed | Web of Science | Google Scholar
  • 179. de Magalhães JP. Cellular senescence in normal physiology. Science 384: 1300–1301, 2024. doi:10.1126/science.adj7050.
    Crossref | PubMed | Web of Science | Google Scholar
  • 180. Kim MH, Kino-Oka M. Bioprocessing strategies for pluripotent stem cells based on Waddington’s epigenetic landscape. Trends Biotechnol 36: 89–104, 2018. doi:10.1016/j.tibtech.2017.10.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 181. Moris N, Pina C, Arias AM. Transition states and cell fate decisions in epigenetic landscapes. Nat Rev Genet 17: 693–703, 2016. doi:10.1038/nrg.2016.98.
    Crossref | PubMed | Web of Science | Google Scholar
  • 182. Rajagopal J, Stanger BZ. Plasticity in the adult: how should the Waddington diagram be applied to regenerating tissues? Dev Cell 36: 133–137, 2016. doi:10.1016/j.devcel.2015.12.021.
    Crossref | PubMed | Web of Science | Google Scholar
  • 183. Watanabe A, Yamada Y, Yamanaka S. Epigenetic regulation in pluripotent stem cells: a key to breaking the epigenetic barrier. Philos Trans R Soc Lond B Biol Sci 368: 20120292, 2013. doi:10.1098/rstb.2012.0292.
    Crossref | PubMed | Web of Science | Google Scholar
  • 184. de Lima Camillo LP, Quinlan RB. A ride through the epigenetic landscape: aging reversal by reprogramming. Geroscience 43: 463–485, 2021. doi:10.1007/s11357-021-00358-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 185. Izadi M, Sadri N, Abdi A, Serajian S, Jalalei D, Tahmasebi S. Epigenetic biomarkers in aging and longevity: current and future application. Life Sci 351: 122842, 2024. doi:10.1016/j.lfs.2024.122842.
    Crossref | PubMed | Web of Science | Google Scholar
  • 186. Wang K, Liu H, Hu Q, Wang L, Liu J, Zheng Z, Zhang W, Ren J, Zhu F, Liu GH. Epigenetic regulation of aging: implications for interventions of aging and diseases. Signal Transduct Target Ther 7: 374, 2022. doi:10.1038/s41392-022-01211-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 187. Debès C, Papadakis A, Groönke S, Karalay O, Tain LS, Mizi A, Nakamura S, Hahn O, Weigelt C, Josipovic N, Zirkel A, Brusius I, Sofiadis K, Lamprousi M, Lu YX, Huang W, Esmaillie R, Kubacki T, Späth MR, Schermer B, Benzing T, Müller RU, Antebi A, Partridge L, Papantonis A, Beyer A. Ageing-associated changes in transcriptional elongation influence longevity. Nature 616: 814–821, 2023. doi:10.1038/s41586-023-05922-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 188. Pal S, Tyler JK. Epigenetics and aging. Sci Adv 2: e1600584, 2016. doi:10.1126/sciadv.1600584.
    Crossref | PubMed | Web of Science | Google Scholar
  • 189. Schumann GG, Fuchs NV, Tristán-Ramos P, Sebe A, Ivics Z, Heras SR. The impact of transposable element activity on therapeutically relevant human stem cells. Mob DNA 10: 9, 2019. doi:10.1186/s13100-019-0151-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 190. Friedmann-Morvinski D, Verma IM. Dedifferentiation and reprogramming: origins of cancer stem cells. EMBO Rep 15: 244–253, 2014. doi:10.1002/embr.201338254.
    Crossref | PubMed | Web of Science | Google Scholar
  • 191. Parras A, Vílchez-Acosta A, Desdín-Micó G, Picó S, Mrabti C, Montenegro-Borbolla E, Maroun CY, Haghani A, Brooke R, Del Carmen Maza M, Rechsteiner C, Battiston F, Branchina C, Perez K, Horvath S, Bertelli C, Sempoux C, Ocampo A. In vivo reprogramming leads to premature death linked to hepatic and intestinal failure. Nat Aging 3: 1509–1520, 2023. doi:10.1038/s43587-023-00528-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 192. Williams BR, Cho JS. Hormone replacement: the fountain of youth? Prim Care 44: 481–498, 2017. doi:10.1016/j.pop.2017.04.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 193. Samaras N, Papadopoulou MA, Samaras D, Ongaro F. Off-label use of hormones as an antiaging strategy: a review. Clin Interv Aging 9: 1175–1186, 2014. doi:10.2147/CIA.S48918.
    Crossref | PubMed | Web of Science | Google Scholar
  • 194. Fraile M, Eiro N, Costa LA, Martín A, Vizoso FJ. Aging and mesenchymal stem cells: basic concepts, challenges and strategies. Biology (Basel) 11, 2022. doi:10.3390/biology11111678.
    Crossref | PubMed | Web of Science | Google Scholar
  • 195. Keyes BE, Fuchs E. Stem cells: aging and transcriptional fingerprints. J Cell Biol 217: 79–92, 2018. doi:10.1083/jcb.201708099.
    Crossref | PubMed | Web of Science | Google Scholar
  • 196. Liu MN, Lan Q, Wu H, Qiu CW. Rejuvenation of young blood on aging organs: effects, circulating factors, and mechanisms. Heliyon 10: e32652, 2024. doi:10.1016/j.heliyon.2024.e32652.
    Crossref | PubMed | Web of Science | Google Scholar
  • 197. Rebo J, Mehdipour M, Gathwala R, Causey K, Liu Y, Conboy MJ, Conboy IM. A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood. Nat Commun 7: 13363, 2016. doi:10.1038/ncomms13363.
    Crossref | PubMed | Web of Science | Google Scholar
  • 198. Kang S, Moser VA, Svendsen CN, Goodridge HS. Rejuvenating the blood and bone marrow to slow aging-associated cognitive decline and Alzheimer’s disease. Commun Biol 3: 69, 2020. doi:10.1038/s42003-020-0797-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 199. Hofmann B. Young blood rejuvenates old bodies: a call for reflection when moving from mice to men. Transfus Med Hemother 45: 67–71, 2018. doi:10.1159/000481828.
    Crossref | PubMed | Web of Science | Google Scholar
  • 200. McCay CM, Crowell MF, Maynard LA. The effect of retarded growth upon the length of life span and upon the ultimate body size. J Nutr 10: 63–79, 1935. doi:10.1093/jn/10.1.63.
    Crossref | Google Scholar
  • 201. McDonald RB, Ramsey JJ. Honoring Clive McCay and 75 years of calorie restriction research. J Nutr 140: 1205–1210, 2010. doi:10.3945/jn.110.122804.
    Crossref | PubMed | Web of Science | Google Scholar
  • 202. Le Bourg E. Predicting whether dietary restriction would increase longevity in species not tested so far. Ageing Res Rev 9: 289–297, 2010. doi:10.1016/j.arr.2010.01.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 203. Fontana L, Partridge L, Longo VD. Extending healthy life span—from yeast to humans. Science 328: 321–326, 2010. doi:10.1126/science.1172539.
    Crossref | PubMed | Web of Science | Google Scholar
  • 204. Kealy RD, Lawler DF, Ballam JM, Mantz SL, Biery DN, Greeley EH, Lust G, Segre M, Smith GK, Stowe HD. Effects of diet restriction on life span and age-related changes in dogs. J Am Vet Med Assoc 220: 1315–1320, 2002. doi:10.2460/javma.2002.220.1315.
    Crossref | PubMed | Web of Science | Google Scholar
  • 205. Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM, Allison DB, Cruzen C, Simmons HA, Kemnitz JW, Weindruch R. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325: 201–204, 2009. doi:10.1126/science.1173635.
    Crossref | PubMed | Web of Science | Google Scholar
  • 206. Mattison JA, Roth GS, Beasley TM, Tilmont EM, Handy AM, Herbert RL, Longo DL, Allison DB, Young JE, Bryant M, Barnard D, Ward WF, Qi W, Ingram DK, de Cabo R. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature 489: 318–321, 2012. doi:10.1038/nature11432.
    Crossref | PubMed | Web of Science | Google Scholar
  • 207. Mattison JA, Colman RJ, Beasley TM, Allison DB, Kemnitz JW, Roth GS, Ingram DK, Weindruch R, de Cabo R, Anderson RM. Caloric restriction improves health and survival of rhesus monkeys. Nat Commun 8: 14063, 2017. doi:10.1038/ncomms14063.
    Crossref | PubMed | Web of Science | Google Scholar
  • 208. Austad SN. Ageing: Mmxed results for dieting monkeys. Nature 489: 210–211, 2012. doi:10.1038/nature11484.
    Crossref | PubMed | Web of Science | Google Scholar
  • 209. Maxmen A. Calorie restriction falters in the long run. Nature 488: 569, 2012. doi:10.1038/488569a.
    Crossref | PubMed | Web of Science | Google Scholar
  • 210. Sohal RS, Forster MJ. Caloric restriction and the aging process: a critique. Free Radic Biol Med 73: 366–382, 2014. doi:10.1016/j.freeradbiomed.2014.05.015.
    Crossref | PubMed | Web of Science | Google Scholar
  • 211. Harper JM, Leathers CW, Austad SN. Does caloric restriction extend life in wild mice? Aging Cell 5: 441–449, 2006. doi:10.1111/j.1474-9726.2006.00236.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 212. Liao CY, Rikke BA, Johnson TE, Diaz V, Nelson JF. Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening. Aging Cell 9: 92–95, 2010. doi:10.1111/j.1474-9726.2009.00533.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 213. Schleit J, Johnson SC, Bennett CF, Simko M, Trongtham N, Castanza A, , et al.. Molecular mechanisms underlying genotype-dependent responses to dietary restriction. Aging Cell 12: 1050–1061, 2013. doi:10.1111/acel.12130.
    Crossref | PubMed | Web of Science | Google Scholar
  • 214. Mitchell SJ, Madrigal-Matute J, Scheibye-Knudsen M, Fang E, Aon M, Gonzalez-Reyes JA, , et al.. Effects of sex, strain, and energy intake on hallmarks of aging in mice. Cell Metab 23: 1093–1112, 2016. doi:10.1016/j.cmet.2016.05.027.
    Crossref | PubMed | Web of Science | Google Scholar
  • 215. Di Francesco A, Deighan AG, Litichevskiy L, Chen Z, Luciano A, Robinson L, Garland G, Donato H, Vincent M, Schott W, Wright KM, Raj A, Prateek GV, Mullis M, Hill WG, Zeidel ML, Peters LL, Harding F, Botstein D, Korstanje R, Thaiss CA, Freund A, Churchill GA. Dietary restriction impacts health and lifespan of genetically diverse mice. Nature 634: 684–692, 2024. doi:10.1038/s41586-024-08026-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 216. Selman C, Swindell WR. Putting a strain on diversity. EMBO J 37: e100862, 2018. doi:10.15252/embj.2018100862.
    Crossref | PubMed | Web of Science | Google Scholar
  • 217. Spiridonova O, Kriukov D, Nemirovich-Danchenko N, Peshkin L. On standardization of controls in lifespan studies. Aging (Albany NY) 16: 3047–3055, 2024. doi:10.18632/aging.205604.
    Crossref | PubMed | Google Scholar
  • 218. Pabis K, Barardo D, Gruber J, Sirbu O, Malavolta M, Selvarajoo K, Kaeberlein M, Kennedy BK. The impact of short-lived controls on the interpretation of lifespan experiments and progress in geroscience - through the lens of the “900-day rule.” Ageing Res Rev 101: 102412, 2024. doi:10.1016/j.arr.2024.102512.
    Google Scholar
  • 219. Hofer SJ, Carmona-Gutierrez D, Mueller MI, Madeo F. The ups and downs of caloric restriction and fasting: from molecular effects to clinical application. EMBO Mol Med 14: e14418, 2022. doi:10.15252/emmm.202114418.
    Crossref | PubMed | Web of Science | Google Scholar
  • 220. Dakic T, Jevdjovic T, Vujovic P, Mladenovic A. The less we eat, the longer we live: can caloric restriction help us become centenarians? Int J Mol Sci 23: 6546, 2022. doi:10.3390/ijms23126546.
    Crossref | PubMed | Web of Science | Google Scholar
  • 221. Eissenberg JC. Hungering for immortality. Mo Med 115: 12–17, 2018.
    PubMed | Google Scholar
  • 222. Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev 39: 36–45, 2017. doi:10.1016/j.arr.2016.08.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 223. Huffman KM, Parker DC, Bhapkar M, Racette SB, Martin CK, Redman LM, Das SK, Connelly MA, Pieper CF, Orenduff M, Ross LM, Ramaker ME, Dorling JL, Rosen CJ, Shalaurova I, Otvos JD, Kraus VB, Kraus WE; CALERIE™ Investigators. Calorie restriction improves lipid-related emerging cardiometabolic risk factors in healthy adults without obesity: Distinct influences of BMI and sex from CALERIE a multicentre, phase 2, randomised controlled trial. EClinicalMedicine 43: 101261, 2022. doi:10.1016/j.eclinm.2021.101261.
    Crossref | PubMed | Web of Science | Google Scholar
  • 224. Hong J, Yun CO. Telomere gene therapy: polarizing therapeutic goals for treatment of various diseases. Cells 8, 2019. doi:10.3390/cells8050392.
    Crossref | PubMed | Web of Science | Google Scholar
  • 225. Borges G, Criqui M, Harrington L. Tieing together loose ends: telomere instability in cancer and aging. Mol Oncol 16: 3380–3396, 2022. doi:10.1002/1878-0261.13299.
    Crossref | PubMed | Web of Science | Google Scholar
  • 226. DeBoy EA, Tassia MG, Schratz KE, Yan SM, Cosner ZL, McNally EJ, Gable DL, Xiang Z, Lombard DB, Antonarakis ES, Gocke CD, McCoy RC, Armanios M. Familial clonal hematopoiesis in a long telomere syndrome. N Engl J Med 388: 2422–2433, 2023. doi:10.1056/NEJMoa2300503.
    Crossref | PubMed | Web of Science | Google Scholar
  • 227. McNally EJ, Luncsford PJ, Armanios M. Long telomeres and cancer risk: the price of cellular immortality. J Clin Invest 129: 3474–3481, 2019. doi:10.1172/JCI120851.
    Crossref | PubMed | Web of Science | Google Scholar
  • 228. Ertl HC. Immunogenicity and toxicity of AAV gene therapy. Front Immunol 13: 975803, 2022. doi:10.3389/fimmu.2022.975803.
    Crossref | PubMed | Web of Science | Google Scholar
  • 229. Mücke MM, Fong S, Foster GR, Lillicrap D, Miesbach W, Zeuzem S. Adeno-associated viruses for gene therapy—clinical implications and liver-related complications, a guide for hepatologists. J Hepatol 80: 352–361, 2024. doi:10.1016/j.jhep.2023.10.029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 230. Swain F. Inside the Secretive Life-Extension Clinic. WIRED, 2025. https://www.wired.com/story/bioviva-gene-therapies-liz-parrish-longevity/ [2025 Jan 30].
    Google Scholar
  • 231. GBD 2021 Risk Factors Collaborators. Global burden and strength of evidence for 88 risk factors in 204 countries and 811 subnational locations, 1990-2021: a systematic analysis for the Global Burden of Disease Study 2021. Lancet 403: 2162–2203, 2024. doi:10.1016/S0140-6736(24)00933-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 232. Bosnes I, Nordahl HM, Stordal E, Bosnes O, Myklebust TA, Almkvist O. Lifestyle predictors of successful aging: a 20-year prospective HUNT study. PLoS One 14: e0219200, 2019. doi:10.1371/journal.pone.0219200.
    Crossref | PubMed | Web of Science | Google Scholar
  • 233. Jamison DT, Summers LH, Chang AY, Karlsson O, Mao W, Norheim OF, , et al.. Global health 2050: the path to halving premature death by mid-century. Lancet 404: 1561–1614, 2024. doi:10.1016/S0140-6736(24)01439-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 234. Bian Z, Wang L, Fan R, Sun J, Yu L, Xu M, Timmers P, Shen X, Wilson JF, Theodoratou E, Wu X, Li X. Genetic predisposition, modifiable lifestyles, and their joint effects on human lifespan: evidence from multiple cohort studies. BMJ Evid Based Med 29: 255–263, 2024. doi:10.1136/bmjebm-2023-112583.
    Crossref | PubMed | Web of Science | Google Scholar
  • 235. Argentieri MA, Amin N, Nevado-Holgado AJ, Sproviero W, Collister JA, Keestra SM, Kuilman MM, Ginos BN, Ghanbari M, Doherty A, Hunter DJ, Alvergne A, van Duijn CM. Integrating the environmental and genetic architectures of aging and mortality. Nat Med 31: 1016–1025, 2025. doi:10.1038/s41591-024-03483-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 236. González-Rodríguez P, Füllgrabe J, Joseph B. The hunger strikes back: an epigenetic memory for autophagy. Cell Death Differ 30: 1404–1415, 2023. doi:10.1038/s41418-023-01159-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 237. Wu G, Zhang X, Gao F. The epigenetic landscape of exercise in cardiac health and disease. J Sport Health Sci 10: 648–659, 2021. doi:10.1016/j.jshs.2020.12.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 238. Plaza-Diaz J, Izquierdo D, Torres-Martos A, Baig AT, Aguilera CM, Ruiz-Ojeda FJ. Impact of physical activity and exercise on the epigenome in skeletal muscle and effects on systemic metabolism. Biomedicines 10: 126, 2022. doi:10.3390/biomedicines10010126.
    Crossref | PubMed | Web of Science | Google Scholar
  • 239. Abraham MJ, El Sherbini A, El-Diasty M, Askari S, Szewczuk MR. Restoring epigenetic reprogramming with diet and exercise to improve health-related metabolic diseases. Biomolecules 13, 2023. doi:10.3390/biom13020318.
    Crossref | PubMed | Web of Science | Google Scholar
  • 240. Li Y, Pan A, Wang DD, Liu X, Dhana K, Franco OH, Kaptoge S, Di Angelantonio E, Stampfer M, Willett WC, Hu FB. Impact of healthy lifestyle factors on life expectancies in the US population. Circulation 138: 345–355, 2018. doi:10.1161/CIRCULATIONAHA.117.032047.
    Crossref | PubMed | Web of Science | Google Scholar
  • 241. Nguyen XT, Li Y, Wang DD, Whitbourne SB, Houghton SC, Hu FB, Willett WC, Sun YV, Djousse L, Gaziano JM, Cho K, Wilson PW; VA Million Veteran Program. Impact of 8 lifestyle factors on mortality and life expectancy among United States veterans: The Million Veteran Program. Am J Clin Nutr 119: 127–135, 2024. doi:10.1016/j.ajcnut.2023.10.032.
    Crossref | PubMed | Web of Science | Google Scholar
  • 242. Di Fusco SA, Spinelli A, Castello L, Marino G, Maraschi I, Gulizia MM, Gabrielli D, Colivicchi F. Do pathophysiologic mechanisms linking unhealthy lifestyle to cardiovascular disease and cancer imply shared preventive measures?—a critical narrative review. Circ J 88: 189–197, 2024. doi:10.1253/circj.CJ-21-0459.
    Crossref | PubMed | Web of Science | Google Scholar
  • 243. Islami F, Marlow EC, Thomson B, McCullough ML, Rumgay H, Gapstur SM, Patel AV, Soerjomataram I, Jemal A. Proportion and number of cancer cases and deaths attributable to potentially modifiable risk factors in the United States, 2019. CA Cancer J Clin 74: 405–432, 2024. doi:10.3322/caac.21858.
    Crossref | PubMed | Web of Science | Google Scholar
  • 244. Zhang R, Lu Y, Bian Z, Zhou S, Xu L, Jiang F, Yuan S, Tan X, Chen X, Ding Y, Li X. Sleep, physical activity, and sedentary behaviors in relation to overall cancer and site-specific cancer risk: a prospective cohort study. iScience 27: 109931, 2024. doi:10.1016/j.isci.2024.109931.
    Crossref | PubMed | Web of Science | Google Scholar
  • 245. Li X, Zhu Y, Yan T, Fang J, Xu X, Xu X. Association between C-reactive protein, Life's Essential 8, and mortality in American adults: insights from NHANES 2005-2010 data analysis. Exp Gerontol 196: 112590, 2024. doi:10.1016/j.exger.2024.112590.
    Crossref | PubMed | Web of Science | Google Scholar
  • 246. Langevin HM, Weber W, Chen W. Integrated multicomponent interventions to support healthy aging of the whole person. Aging Cell 23: e14001, 2024. doi:10.1111/acel.14001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 247. Matta K, Viallon V, Botteri E, Peveri G, Dahm C, Nannsen AO, , et al.. Healthy lifestyle change and all-cause and cancer mortality in the European Prospective Investigation into Cancer and Nutrition cohort. BMC Med 22: 210, 2024. doi:10.1186/s12916-024-03362-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 248. Lee CH, Han KD, Kim DH, Kwak MS. Continuing regular physical activity and maintaining body weight have a synergistic interaction in improving survival: a population-based cohort study including 6.5 million people. Eur J Prev Cardiol 29: 547–555, 2022. doi:10.1093/eurjpc/zwab190.
    Crossref | PubMed | Web of Science | Google Scholar
  • 249. Robb C, Carr PR, Ball J, Owen A, Beilin LJ, Newman AB, Nelson MR, Reid CM, Orchard SG, Neumann JT, Tonkin AM, Wolfe R, McNeil JJ. Association of a healthy lifestyle with mortality in older people. BMC Geriatr 23: 646, 2023. doi:10.1186/s12877-023-04247-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 250. Zhang YB, Pan XF, Chen J, Cao A, Xia L, Zhang Y, Wang J, Li H, Liu G, Pan A. Combined lifestyle factors, all-cause mortality and cardiovascular disease: a systematic review and meta-analysis of prospective cohort studies. J Epidemiol Community Health 75: 92–99, 2021. doi:10.1136/jech-2020-214050.
    Crossref | PubMed | Web of Science | Google Scholar
  • 251. Bonaccio M, Di Castelnuovo A, Costanzo S, De Curtis A, Persichillo M, Cerletti C, Donati MB, de Gaetano G, Iacoviello L; Moli-sani Study Investigators. Impact of combined healthy lifestyle factors on survival in an adult general population and in high-risk groups: prospective results from the Moli-sani Study. J Intern Med 286: 207–220, 2019. doi:10.1111/joim.12907.
    Crossref | PubMed | Web of Science | Google Scholar
  • 252. Zhang G, Fu J, Zhang H, Xu X, Cao Z. The impact of Life’s Essentials 8 on sarcopenia prevalence among adults in the United States. Exp Gerontol 198: 112631, 2024. doi:10.1016/j.exger.2024.112631.
    Crossref | PubMed | Web of Science | Google Scholar
  • 253. Naderian M, Norland K, Schaid DJ, Kullo IJ. Development and evaluation of a comprehensive prediction model for incident coronary heart disease using genetic, social, and lifestyle-psychological factors: a prospective analysis of the UK Biobank. Ann Intern Med 178: 1–10, 2025. doi:10.7326/ANNALS-24-00716.
    Crossref | PubMed | Web of Science | Google Scholar
  • 254. Li Y, Wang K, Jigeer G, Jensen G, Tucker KL, Lv Y, Shi X, Gao X. Healthy lifestyle and the likelihood of becoming a centenarian. JAMA Netw Open 7: e2417931, 2024. doi:10.1001/jamanetworkopen.2024.17931.
    Crossref | Web of Science | Google Scholar
  • 255. Livingston G, Huntley J, Liu KY, Costafreda SG, Selbæk G, Alladi S, Ames D, Banerjee S, Burns A, Brayne C, Fox NC, Ferri CP, Gitlin LN, Howard R, Kales HC, Kivimäki M, Larson EB, Nakasujja N, Rockwood K, Samus Q, Shirai K, Singh-Manoux A, Schneider LS, Walsh S, Yao Y, Sommerlad A, Mukadam N. Dementia prevention, intervention, and care: 2024 report of the Lancet standing Commission. Lancet 404: 572–628, 2024. doi:10.1016/S0140-6736(24)01296-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 256. Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, Brayne C, Burns A, Cohen-Mansfield J, Cooper C, Costafreda SG, Dias A, Fox N, Gitlin LN, Howard R, Kales HC, Kivimaki M, Larson EB, Ogunniyi A, Orgeta V, Ritchie K, Rockwood K, Sampson EL, Samus Q, Schneider LS, Selbaek G, Teri L, Mukadam N. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet 396: 413–446, 2020. doi:10.1016/S0140-6736(20)30367-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 257. Sato R, Vatic M, Peixoto da Fonseca GW, Anker SD, von Haehling S. Biological basis and treatment of frailty and sarcopenia. Cardiovasc Res 120: 982–998, 2024. doi:10.1093/cvr/cvae073.
    Crossref | PubMed | Web of Science | Google Scholar
  • 258. Helmrich SP, Ragland DR, Leung RW, Paffenbarger RS Jr. Physical activity and reduced occurrence of non-insulin-dependent diabetes mellitus. N Engl J Med 325: 147–152, 1991. doi:10.1056/NEJM199107183250302.
    Crossref | PubMed | Web of Science | Google Scholar
  • 259. Dove A, Wang J, Huang H, Dunk MM, Sakakibara S, Guitart-Masip M, Papenberg G, Xu W. Diabetes, prediabetes, and brain aging: the role of healthy lifestyle. Diabetes Care 47: 1794–1802, 2024. doi:10.2337/dc24-0860.
    Crossref | PubMed | Web of Science | Google Scholar
  • 260. Farina FR, Bridgeman K, Gregory S, Crivelli L, Foote IF, Jutila OI, , et al.. Next generation brain health: transforming global research and public health to promote prevention of dementia and reduce its risk in young adult populations. Lancet Healthy Longev 5: 100665, 2024. doi:10.1016/j.lanhl.2024.100665.
    Crossref | PubMed | Web of Science | Google Scholar
  • 261. Bushnell C, Kernan WN, Sharrief AZ, Chaturvedi S, Cole JW, Cornwell WK 3rd, Cosby-Gaither C, Doyle S, Goldstein LB, Lennon O, Levine DA, Love M, Miller E, Nguyen-Huynh M, Rasmussen-Winkler J, Rexrode KM, Rosendale N, Sarma S, Shimbo D, Simpkins AN, Spatz ES, Sun LR, Tangpricha V, Turnage D, Velazquez G, Whelton PK. 2024 Guideline for the Primary Prevention of Stroke: a guideline from the American Heart Association/American Stroke Association. Stroke 55: e344–e424, 2024. doi:10.1161/STR.0000000000000475.
    Crossref | PubMed | Web of Science | Google Scholar
  • 262. Lin L, Hu Y, Lei F, Huang X, Zhang X, Sun T, Liu W, Li R, Zhang XJ, Cai J, She ZG, Wang G, Li H. Cardiovascular health and cancer mortality: evidence from US NHANES and UK Biobank cohort studies. BMC Med 22: 368, 2024. doi:10.1186/s12916-024-03553-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 263. Sawalla Guseh J, Lieberman D, Baggish A. The evidence for exercise in medicine—a new review series. NEJM Evid 1: EVIDra2100002, 2022. doi:10.1056/EVIDra2100002.
    Crossref | PubMed | Google Scholar
  • 264. Handschin C, Spiegelman BM. The role of exercise and PGC1alpha in inflammation and chronic disease. Nature 454: 463–469, 2008. doi:10.1038/nature07206.
    Crossref | PubMed | Web of Science | Google Scholar
  • 265. Jiang G, Zhang W, Kang H, Wang J, Liu Z, Wang Z, Huang D, Gao A. The association between weekly exercise patterns and acceleration of aging: evidence from a population-based study. Prev Med 187: 108091, 2024. doi:10.1016/j.ypmed.2024.108091.
    Crossref | PubMed | Web of Science | Google Scholar
  • 266. Löllgen H, Böckenhoff A, Knapp G. Physical activity and all-cause mortality: an updated meta-analysis with different intensity categories. Int J Sports Med 30: 213–224, 2009. doi:10.1055/s-0028-1128150.
    Crossref | PubMed | Web of Science | Google Scholar
  • 267. Reimers CD, Knapp G, Reimers AK. Does physical activity increase life expectancy? A review of the literature. J Aging Res 2012: 243958, 2012. doi:10.1155/2012/243958.
    Crossref | PubMed | Google Scholar
  • 268. Mori I, Ishizuka T, Morita H, Matsumoto M, Uno Y, Kajita K, Ikeda T, Fujioka K, Matsubara K. Comparison of biochemical data, blood pressure and physical activity between longevity and non-longevity districts in Japan. Circ J 72: 1680–1684, 2008. doi:10.1253/circj.cj-07-1081.
    Crossref | PubMed | Web of Science | Google Scholar
  • 269. Chudasama YV, Khunti KK, Zaccardi F, Rowlands AV, Yates T, Gillies CL, Davies MJ, Dhalwani NN. Physical activity, multimorbidity, and life expectancy: a UK Biobank longitudinal study. BMC Med 17: 108, 2019. doi:10.1186/s12916-019-1339-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 270. Buchman AS, Yu L, Boyle PA, Shah RC, Bennett DA. Total daily physical activity and longevity in old age. Arch Intern Med 172: 444–446, 2012. doi:10.1001/archinternmed.2011.1477.
    Crossref | PubMed | Web of Science | Google Scholar
  • 271. Brandts L, van den Brandt PA. Body size, non-occupational physical activity and the chance of reaching longevity in men and women: findings from the Netherlands Cohort Study. J Epidemiol Community Health 73: 239–249, 2019. doi:10.1136/jech-2018-211410.
    Crossref | PubMed | Web of Science | Google Scholar
  • 272. Altulea A, Rutten MG, Verdijk LB, Demaria M. Sport and longevity: an observational study of international athletes. Geroscience, 2024. doi:10.1007/s11357-024-01307-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 273. Zaccardi F, Rowlands AV, Dempsey PC, Razieh C, Henson J, Goldney J, Maylor BD, Bhattacharjee A, Chudasama Y, Edwardson C, Laukkanen JA, Ekelund U, Davies MJ, Khunti K, Yates T. Interplay between physical activity volume and intensity with modeled life expectancy in women and men: a prospective cohort analysis. J Sport Health Sci 14: 100970, 2024. doi:10.1016/j.jshs.2024.100970.
    Crossref | PubMed | Web of Science | Google Scholar
  • 274. Lee IM, Skerrett PJ. Physical activity and all-cause mortality: what is the dose-response relation? Med Sci Sports Exerc 33: S459–S471, 2001. doi:10.1097/00005768-200106001-00016.
    Crossref | PubMed | Web of Science | Google Scholar
  • 275. Lear SA, Hu W, Rangarajan S, Gasevic D, Leong D, Iqbal R, Casanova A, Swaminathan S, Anjana RM, Kumar R, Rosengren A, Wei L, Yang W, Chuangshi W, Huaxing L, Nair S, Diaz R, Swidon H, Gupta R, Mohammadifard N, Lopez-Jaramillo P, Oguz A, Zatonska K, Seron P, Avezum A, Poirier P, Teo K, Yusuf S. The effect of physical activity on mortality and cardiovascular disease in 130 000 people from 17 high-income, middle-income, and low-income countries: the PURE study. Lancet 390: 2643–2654, 2017. doi:10.1016/S0140-6736(17)31634-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 276. López-Bueno R, Ahmadi M, Stamatakis E, Yang L, Del Pozo Cruz B. Prospective associations of different combinations of aerobic and muscle-strengthening activity with all-cause, cardiovascular, and cancer mortality. JAMA Intern Med 183: 982–990, 2023. doi:10.1001/jamainternmed.2023.3093.
    Crossref | PubMed | Web of Science | Google Scholar
  • 277. Schwendinger F, Infanger D, Lichtenstein E, Hinrichs T, Knaier R, Rowlands AV, Schmidt-Trucksäss A. Intensity or volume: the role of physical activity in longevity. Eur J Prev Cardiol 32: 10–19, 2025. doi:10.1093/eurjpc/zwae295.
    Crossref | PubMed | Web of Science | Google Scholar
  • 278. Lee IM, Paffenbarger RS Jr. Associations of light, moderate, and vigorous intensity physical activity with longevity. The Harvard Alumni Health Study. Am J Epidemiol 151: 293–299, 2000. doi:10.1093/oxfordjournals.aje.a010205.
    Crossref | PubMed | Web of Science | Google Scholar
  • 279. Roberts WC. An agent with lipid-lowering, antihypertensive, positive inotropic, negative chronotropic, vasodilating, diuretic, anorexigenic, weight-reducing, cathartic, hypoglycemic, tranquilizing, hypnotic and antidepressive qualities. Am J Cardiol 53: 261–262, 1984. doi:10.1016/0002-9149(84)90725-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 280. Veerman L, Tarp J, Wijaya R, Wanjau MN, Möller H, Haigh F, Lucas P, Milat A. Physical activity and life expectancy: a life-table analysis. Br J Sports Med 59: 333–338, 2025. doi:10.1136/bjsports-2024-108125.
    Crossref | PubMed | Web of Science | Google Scholar
  • 281. Ruegsegger GN, Booth FW. Health benefits of exercise. Cold Spring Harb Perspect Med 8: a029694, 2018. doi:10.1101/cshperspect.a029694.
    Crossref | PubMed | Web of Science | Google Scholar
  • 282. Hsu B, Merom D, Blyth FM, Naganathan V, Hirani V, Le Couteur DG, Seibel MJ, Waite LM, Handelsman DJ, Cumming RG. Total physical activity, exercise intensity, and walking speed as predictors of all-cause and cause-specific mortality over 7 years in older men: the Concord Health and Aging in Men Project. J Am Med Dir Assoc 19: 216–222, 2018. doi:10.1016/j.jamda.2017.08.018.
    Crossref | PubMed | Web of Science | Google Scholar
  • 283. Lee IM, Hsieh CC, Paffenbarger RS Jr. Exercise intensity and longevity in men. The Harvard Alumni Health Study. JAMA 273: 1179–1184, 1995. doi:10.1001/jama.1995.03520390039030.
    Crossref | PubMed | Web of Science | Google Scholar
  • 284. Saint-Maurice PF, Graubard BI, Troiano RP, Berrigan D, Galuska DA, Fulton JE, Matthews CE. Estimated number of deaths prevented through increased physical activity among US adults. JAMA Intern Med 182: 349–352, 2022. doi:10.1001/jamainternmed.2021.7755.
    Crossref | PubMed | Web of Science | Google Scholar
  • 285. Powell KE, Thompson PD, Caspersen CJ, Kendrick JS. Physical activity and the incidence of coronary heart disease. Annu Rev Public Health 8: 253–287, 1987. doi:10.1146/annurev.pu.08.050187.001345.
    Crossref | PubMed | Web of Science | Google Scholar
  • 286. Nocon M, Hiemann T, Müller-Riemenschneider F, Thalau F, Roll S, Willich SN. Association of physical activity with all-cause and cardiovascular mortality: a systematic review and meta-analysis. Eur J Cardiovasc Prev Rehabil 15: 239–246, 2008. doi:10.1097/HJR.0b013e3282f55e09.
    Crossref | PubMed | Web of Science | Google Scholar
  • 287. Sattelmair J, Pertman J, Ding EL, Kohl HW 3rd, Haskell W, Lee IM. Dose response between physical activity and risk of coronary heart disease: a meta-analysis. Circulation 124: 789–795, 2011. doi:10.1161/CIRCULATIONAHA.110.010710.
    Crossref | PubMed | Web of Science | Google Scholar
  • 288. Jeong SW, Kim SH, Kang SH, Kim HJ, Yoon CH, Youn TJ, Chae IH. Mortality reduction with physical activity in patients with and without cardiovascular disease. Eur Heart J 40: 3547–3555, 2019. doi:10.1093/eurheartj/ehz564.
    Crossref | PubMed | Web of Science | Google Scholar
  • 289. Lee JY, Ryu S, Cheong E, Sung KC. Association of physical activity and inflammation with all-cause, cardiovascular-related, and cancer-related mortality. Mayo Clin Proc 91: 1706–1716, 2016. doi:10.1016/j.mayocp.2016.08.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 290. Martinez-Gomez D, Luo M, Huang Y, Rodríguez-Artalejo F, Ekelund U, Sotos-Prieto M, Ding D, Lao XQ, Cabanas-Sánchez V. Physical activity and all-cause mortality by age in 4 multinational megacohorts. JAMA Netw Open 7: e2446802, 2024. doi:10.1001/jamanetworkopen.2024.46802.
    Crossref | PubMed | Web of Science | Google Scholar
  • 291. Oguma Y, Sesso HD, Paffenbarger RS Jr, Lee IM. Physical activity and all cause mortality in women: a review of the evidence. Br J Sports Med 36: 162–172, 2002. doi:10.1136/bjsm.36.3.162.
    Crossref | PubMed | Web of Science | Google Scholar
  • 292. Kushi LH, Fee RM, Folsom AR, Mink PJ, Anderson KE, Sellers TA. Physical activity and mortality in postmenopausal women. JAMA 277: 1287–1292, 1997.
    Crossref | PubMed | Web of Science | Google Scholar
  • 293. Tipton CM. The history of “Exercise Is Medicine” in ancient civilizations. Adv Physiol Educ 38: 109–117, 2014. doi:10.1152/advan.00136.2013.
    Link | Web of Science | Google Scholar
  • 294. Lavin KM, Coen PM, Baptista LC, Bell MB, Drummer D, Harper SA, Lixandrão ME, McAdam JS, O’Bryan SM, Ramos S, Roberts LM, Vega RB, Goodpaster BH, Bamman MM, Buford TW. State of knowledge on molecular adaptations to exercise in humans: historical perspectives and future directions. Compr Physiol 12: 3193–3279, 2022. doi:10.1002/cphy.c200033.
    Crossref | PubMed | Web of Science | Google Scholar
  • 295. Furrer R, Hawley JA, Handschin C. The molecular athlete: exercise physiology from mechanisms to medals. Physiol Rev 103: 1693–1787, 2023. doi:10.1152/physrev.00017.2022.
    Link | Web of Science | Google Scholar
  • 296. Qiu Y, Fernández-García B, Lehmann HI, Li G, Kroemer G, López-Otín C, Xiao J. Exercise sustains the hallmarks of health. J Sport Health Sci 12: 8–35, 2023. doi:10.1016/j.jshs.2022.10.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 297. Rebelo-Marques A, De Sousa Lages A, Andrade R, Ribeiro CF, Mota-Pinto A, Carrilho F, Espregueira-Mendes J. Aging hallmarks: the benefits of physical exercise. Front Endocrinol (Lausanne) 9: 258, 2018. doi:10.3389/fendo.2018.00258.
    Crossref | PubMed | Web of Science | Google Scholar
  • 298. Garatachea N, Pareja-Galeano H, Sanchis-Gomar F, Santos-Lozano A, Fiuza-Luces C, Morán M, Emanuele E, Joyner MJ, Lucia A. Exercise attenuates the major hallmarks of aging. Rejuvenation Res 18: 57–89, 2015. doi:10.1089/rej.2014.1623.
    Crossref | PubMed | Web of Science | Google Scholar
  • 299. Goh J, Wong E, Soh J, Maier AB, Kennedy BK. Targeting the molecular & cellular pillars of human aging with exercise. FEBS J 290: 649–668, 2023. doi:10.1111/febs.16337.
    Crossref | PubMed | Web of Science | Google Scholar
  • 300. Lefferts WK, Davis MM, Valentine RJ. Exercise as an aging mimetic: a new perspective on the mechanisms behind exercise as preventive medicine against age-related chronic disease. Front Physiol 13: 866792, 2022. doi:10.3389/fphys.2022.866792.
    Crossref | PubMed | Web of Science | Google Scholar
  • 301. Franklin BA. Preventing exercise-related cardiovascular events: is a medical examination more urgent for physical activity or inactivity? Circulation 129: 1081–1084, 2014. doi:10.1161/CIRCULATIONAHA.114.007641.
    Crossref | PubMed | Web of Science | Google Scholar
  • 302. Pedersen BK, Saltin B. Exercise as medicine—evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand J Med Sci Sports 25, Suppl 3: 1–72, 2015. doi:10.1111/sms.12581.
    Crossref | PubMed | Web of Science | Google Scholar
  • 303. Pedersen BK. The physiology of optimizing health with a focus on exercise as medicine. Annu Rev Physiol 81: 607–627, 2019. doi:10.1146/annurev-physiol-020518-114339.
    Crossref | PubMed | Web of Science | Google Scholar
  • 304. Naci H, Ioannidis JP. Comparative effectiveness of exercise and drug interventions on mortality outcomes: metaepidemiological study. BMJ 347: f5577, 2013. doi:10.1136/bmj.f5577.
    Crossref | PubMed | Web of Science | Google Scholar
  • 305. Booth FW, Roberts CK, Thyfault JP, Ruegsegger GN, Toedebusch RG. Role of inactivity in chronic diseases: evolutionary insight and pathophysiological mechanisms. Physiol Rev 97: 1351–1402, 2017. doi:10.1152/physrev.00019.2016.
    Link | Web of Science | Google Scholar
  • 306. Diniz DG, Bento-Torres J, da Costa VO, Carvalho JP, Tomás AM, Galdino de Oliveira TC, Soares FC, de Macedo L, Jardim NY, Bento-Torres NV, Anthony DC, Brites D, Picanço Diniz CW. The hidden dangers of sedentary living: insights into molecular, cellular, and systemic mechanisms. Int J Mol Sci 25: 10757, 2024. doi:10.3390/ijms251910757.
    Crossref | PubMed | Web of Science | Google Scholar
  • 307. Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN, Katzmarzyk PT; Lancet Physical Activity Series Working Group. Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy. Lancet 380: 219–229, 2012. doi:10.1016/S0140-6736(12)61031-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 308. Wen CP, Wu X. Stressing harms of physical inactivity to promote exercise. Lancet 380: 192–193, 2012. doi:10.1016/S0140-6736(12)60954-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 309. Blair SN, Kohl HW, Gordon NF, Paffenbarger RS Jr. How much physical activity is good for health? Annu Rev Public Health 13: 99–126, 1992. doi:10.1146/annurev.pu.13.050192.000531.
    Crossref | PubMed | Web of Science | Google Scholar
  • 310. Shiroma EJ, Lee IM. Can we proceed with physical activity recommendations if (almost) no clinical trial data exist on mortality? Br J Sports Med 52: 888–889, 2018. doi:10.1136/bjsports-2018-099185.
    Crossref | PubMed | Web of Science | Google Scholar
  • 311. English LK, Ard JD, Bailey RL, Bates M, Bazzano LA, Boushey CJ, Brown C, Butera G, Callahan EH, de Jesus J, Mattes RD, Mayer-Davis EJ, Novotny R, Obbagy JE, Rahavi EB, Sabate J, Snetselaar LG, Stoody EE, Van Horn LV, Venkatramanan S, Heymsfield SB. Evaluation of dietary patterns and all-cause mortality: a systematic review. JAMA Netw Open 4: e2122277, 2021. doi:10.1001/jamanetworkopen.2021.22277.
    Crossref | PubMed | Web of Science | Google Scholar
  • 312. Jayanama K, Theou O, Godin J, Cahill L, Shivappa N, Hébert JR, Wirth MD, Park YM, Fung TT, Rockwood K. Relationship between diet quality scores and the risk of frailty and mortality in adults across a wide age spectrum. BMC Med 19: 64, 2021. doi:10.1186/s12916-021-01918-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 313. Brandhorst S, Longo VD. Protein quantity and source, fasting-mimicking diets, and longevity. Adv Nutr 10: S340–S350, 2019. doi:10.1093/advances/nmz079.
    Crossref | PubMed | Web of Science | Google Scholar
  • 314. Ardisson Korat AV, Shea MK, Jacques PF, Sebastiani P, Wang M, Eliassen AH, Willett WC, Sun Q. Dietary protein intake in midlife in relation to healthy aging—results from the prospective Nurses’ Health Study cohort. Am J Clin Nutr 119: 271–282, 2024. doi:10.1016/j.ajcnut.2023.11.010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 315. Deane CS, Cox J, Atherton PJ. Critical variables regulating age-related anabolic responses to protein nutrition in skeletal muscle. Front Nutr 11: 1419229, 2024. doi:10.3389/fnut.2024.1419229.
    Crossref | PubMed | Web of Science | Google Scholar
  • 316. Krok-Schoen JL, Archdeacon Price A, Luo M, Kelly OJ, Taylor CA. Low dietary protein intakes and associated dietary patterns and functional limitations in an aging population: a NHANES analysis. J Nutr Health Aging 23: 338–347, 2019. doi:10.1007/s12603-019-1174-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 317. Kitada M, Ogura Y, Monno I, Koya D. The impact of dietary protein intake on longevity and metabolic health. EBioMedicine 43: 632–640, 2019. doi:10.1016/j.ebiom.2019.04.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 318. Coelho-Junior HJ, Rodrigues B, Uchida M, Marzetti E. Low protein intake is associated with frailty in older adults: a systematic review and meta-analysis of observational studies. Nutrients 10: 1334, 2018. doi:10.3390/nu10091334.
    Crossref | PubMed | Web of Science | Google Scholar
  • 319. Han M, Woo K, Kim K. Association of protein intake with sarcopenia and related indicators among Korean older adults: a systematic review and meta-analysis. Nutrients 16: 4350, 2024. doi:10.3390/nu16244350.
    Crossref | Web of Science | Google Scholar
  • 320. Carballo-Casla A, Avesani CM, Beridze G, Ortolá R, Garcia-Esquinas E, Lopez-Garcia E, Dai L, Dunk MM, Stenvinkel P, Lindholm B, Carrero JJ, Rodríguez-Artalejo F, Vetrano DL, Calderón-Larrañaga A. Protein intake and mortality in older adults with chronic kidney disease. JAMA Netw Open 7: e2426577, 2024. doi:10.1001/jamanetworkopen.2024.26577.
    Crossref | PubMed | Web of Science | Google Scholar
  • 321. Strasser B, Volaklis K, Fuchs D, Burtscher M. Role of dietary protein and muscular fitness on longevity and aging. Aging Dis 9: 119–132, 2018. doi:10.14336/AD.2017.0202.
    Crossref | PubMed | Web of Science | Google Scholar
  • 322. Hu FB. Diet strategies for promoting healthy aging and longevity: an epidemiological perspective. J Intern Med 295: 508–531, 2024. doi:10.1111/joim.13728.
    Crossref | PubMed | Web of Science | Google Scholar
  • 323. Berger MM, Shenkin A, Schweinlin A, Amrein K, Augsburger M, Biesalski HK, Bischoff SC, Casaer MP, Gundogan K, Lepp HL, de Man AM, Muscogiuri G, Pietka M, Pironi L, Rezzi S, Cuerda C. ESPEN micronutrient guideline. Clin Nutr 41: 1357–1424, 2022. doi:10.1016/j.clnu.2022.02.015.
    Crossref | PubMed | Web of Science | Google Scholar
  • 324. González-Gil EM, Matta M, Morales Berstein F, Cairat M, Nicolas G, Blanco J, , et al.. Associations between degree of food processing and all-cause and cause-specific mortality: a multicentre prospective cohort analysis in 9 European countries. Lancet Reg Health Eur 50: 101208, 2025. doi:10.1016/j.lanepe.2024.101208.
    Crossref | PubMed | Web of Science | Google Scholar
  • 325. de Oliveira Otto MC, Anderson CA, Dearborn JL, Ferranti EP, Mozaffarian D, Rao G, Wylie-Rosett J, Lichtenstein AH; American Heart Association Behavioral Change for Improving Health Factors Committee of the Council on Lifestyle and Cardiometabolic Health and Council on Epidemiology and Prevention, Council on Cardiovascular and Stroke Nursing, Council on Clinical Cardiology, Stroke Council. Dietary Diversity: Implications for Obesity Prevention in Adult Populations: a science advisory from the American Heart Association. Circulation 138: e160–e168, 2018. doi:10.1161/CIR.0000000000000595.
    Crossref | PubMed | Web of Science | Google Scholar
  • 326. Snetselaar LG, de Jesus JM, DeSilva DM, Stoody EE. Dietary Guidelines for Americans, 2020-2025: understanding the scientific process, guidelines, and key recommendations. Nutr Today 56: 287–295, 2021. doi:10.1097/NT.0000000000000512.
    Crossref | PubMed | Google Scholar
  • 327. Venkatesan P. Food is medicine: clinical trials show the health benefits of dietary interventions. Nat Med 30: 916–919, 2024. doi:10.1038/s41591-024-02891-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 328. Dmitrieva NI, Gagarin A, Liu D, Wu CO, Boehm M. Middle-age high normal serum sodium as a risk factor for accelerated biological aging, chronic diseases, and premature mortality. EBioMedicine 87: 104404, 2023. doi:10.1016/j.ebiom.2022.104404.
    Crossref | PubMed | Web of Science | Google Scholar
  • 329. Fadnes LT, Celis-Morales C, Økland JM, Parra-Soto S, Livingstone KM, Ho FK, Pell JP, Balakrishna R, Javadi Arjmand E, Johansson KA, Haaland OA, Mathers JC. Life expectancy can increase by up to 10 years following sustained shifts towards healthier diets in the United Kingdom. Nat Food 4: 961–965, 2023. doi:10.1038/s43016-023-00868-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 330. Fadnes LT, Økland JM, Haaland OA, Johansson KA. Estimating impact of food choices on life expectancy: a modeling study. PLoS Med 19: e1003889, 2022. doi:10.1371/journal.pmed.1003889.
    Crossref | PubMed | Web of Science | Google Scholar
  • 331. Moradell A, Casajús JA, Moreno LA, Vicente-Rodríguez G. Perspectives on diet and exercise interaction for healthy aging: opportunities to reduce malnutrition risk and optimize fitness. Nutrients 17: 596, 2025. doi:10.3390/nu17030596.
    Crossref | PubMed | Web of Science | Google Scholar
  • 332. Rodgers GP, Collins FS. Precision nutrition—the answer to “what to eat to stay healthy”. JAMA 324: 735–736, 2020. doi:10.1001/jama.2020.13601.
    Crossref | PubMed | Web of Science | Google Scholar
  • 333. Bailey RL, Stover PJ. Precision nutrition: the hype is exceeding the science and evidentiary standards needed to inform public health recommendations for prevention of chronic disease. Annu Rev Nutr 43: 385–407, 2023. doi:10.1146/annurev-nutr-061021-025153.
    Crossref | PubMed | Web of Science | Google Scholar
  • 334. Offord C. Misreported meals skew nutrition research data. Science 387: 352, 2025. doi:10.1126/science.adw1659.
    Crossref | PubMed | Google Scholar
  • 335. Stubbs RJ, Hopkins M. Predictive equation helps estimate misreporting of energy intakes in dietary surveys. Nat Food 6: 8–9, 2025. doi:10.1038/s43016-024-01090-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 336. Hirshkowitz M, Whiton K, Albert SM, Alessi C, Bruni O, DonCarlos L, Hazen N, Herman J, Adams Hillard PJ, Katz ES, Kheirandish-Gozal L, Neubauer DN, O’Donnell AE, Ohayon M, Peever J, Rawding R, Sachdeva RC, Setters B, Vitiello MV, Ware JC. National Sleep Foundation’s updated sleep duration recommendations: final report. Sleep Health 1: 233–243, 2015. doi:10.1016/j.sleh.2015.10.004.
    Crossref | PubMed | Google Scholar
  • 337. Chaput JP, Dutil C, Featherstone R, Ross R, Giangregorio L, Saunders TJ, Janssen I, Poitras VJ, Kho ME, Ross-White A, Zankar S, Carrier J. Sleep timing, sleep consistency, and health in adults: a systematic review. Appl Physiol Nutr Metab 45: S232–S247, 2020. doi:10.1139/apnm-2020-0032.
    Crossref | PubMed | Web of Science | Google Scholar
  • 338. Bliwise DL, Young TB. The parable of parabola: what the U-shaped curve can and cannot tell us about sleep. Sleep 30: 1614–1615, 2007. doi:10.1093/sleep/30.12.1614.
    Crossref | PubMed | Web of Science | Google Scholar
  • 339. Hossin MZ. From habitual sleep hours to morbidity and mortality: existing evidence, potential mechanisms, and future agenda. Sleep Health 2: 146–153, 2016. doi:10.1016/j.sleh.2016.01.006.
    Crossref | PubMed | Google Scholar
  • 340. Garbarino S, Lanteri P, Durando P, Magnavita N, Sannita WG. Co-morbidity, mortality, quality of life and the healthcare/welfare/social costs of disordered sleep: a rapid review. Int J Environ Res Public Health 13, 2016. doi:10.3390/ijerph13080831.
    Crossref | PubMed | Web of Science | Google Scholar
  • 341. Zheng NS, Annis J, Master H, Han L, Gleichauf K, Ching JH, Nasser M, Coleman P, Desine S, Ruderfer DM, Hernandez J, Schneider LD, Brittain EL. Sleep patterns and risk of chronic disease as measured by long-term monitoring with commercial wearable devices in the All of Us Research Program. Nat Med 30: 2648–2656, 2024. doi:10.1038/s41591-024-03155-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 342. Cappuccio FP, D'Elia L, Strazzullo P, Miller MA. Sleep duration and all-cause mortality: a systematic review and meta-analysis of prospective studies. Sleep 33: 585–592, 2010. doi:10.1093/sleep/33.5.585.
    Crossref | PubMed | Web of Science | Google Scholar
  • 343. Li H, Qian F, Han L, Feng W, Zheng D, Guo X, Zhang H. Association of healthy sleep patterns with risk of mortality and life expectancy at age of 30 years: a population-based cohort study. QJM 117: 177–186, 2024. doi:10.1093/qjmed/hcad237.
    Crossref | PubMed | Web of Science | Google Scholar
  • 344. Robbins R, Quan SF, Weaver MD, Bormes G, Barger LK, Czeisler CA. Examining sleep deficiency and disturbance and their risk for incident dementia and all-cause mortality in older adults across 5 years in the United States. Aging (Albany NY) 13: 3254–3268, 2021. doi:10.18632/aging.202591.
    Crossref | PubMed | Web of Science | Google Scholar
  • 345. Itani O, Jike M, Watanabe N, Kaneita Y. Short sleep duration and health outcomes: a systematic review, meta-analysis, and meta-regression. Sleep Med 32: 246–256, 2017. doi:10.1016/j.sleep.2016.08.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 346. Åkerstedt T, Narusyte J, Alexanderson K, Svedberg P. Sleep duration, mortality, and heredity-a prospective twin study. Sleep 40: zsx135, 2017. doi:10.1093/sleep/zsx135.
    Crossref | Web of Science | Google Scholar
  • 347. Kohn S, Diament A, Godneva A, Dhir R, Weinberger A, Reisner Y, Rossman H, Segal E. Phenome-wide associations of sleep characteristics in the Human Phenotype Project. Nat Med 31: 1026–1037, 2025. doi:10.1038/s41591-024-03481-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 348. Zhu H, Qin S, Wu M. Association between weekend catch-up sleep and cardiovascular disease: evidence from the National Health and Nutrition Examination Surveys 2017-2018. Sleep Health 10: 98–103, 2024. doi:10.1016/j.sleh.2023.09.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 349. Hirohama K, Imura T, Hori T, Deguchi N, Mitsutake T, Tanaka R. The effects of nonpharmacological sleep hygiene on sleep quality in nonelderly individuals: a systematic review and network meta-analysis of randomized controlled trials. PLoS One 19: e0301616, 2024. doi:10.1371/journal.pone.0301616.
    Crossref | PubMed | Web of Science | Google Scholar
  • 350. Dolezal BA, Neufeld EV, Boland DM, Martin JL, Cooper CB. Interrelationship between sleep and exercise: a systematic review. Adv Prev Med 2017: 1364387, 2017. doi:10.1155/2017/1364387.
    Crossref | PubMed | Google Scholar
  • 351. Giannaki CD, Sakkas GK, Hadjigeorgiou GM, Manconi M, Bargiotas P. Unfolding the role of exercise in the management of sleep disorders. Eur J Appl Physiol 124: 2547–2560, 2024. doi:10.1007/s00421-024-05556-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 352. Windred DP, Burns AC, Lane JM, Olivier P, Rutter MK, Saxena R, Phillips AJ, Cain SW. Brighter nights and darker days predict higher mortality risk: a prospective analysis of personal light exposure in >88,000 individuals. Proc Natl Acad Sci USA 121: e2405924121, 2024. doi:10.1073/pnas.2405924121.
    Crossref | PubMed | Web of Science | Google Scholar
  • 353. Hayat SA, Luben R, Dalzell N, Moore S, Hogervorst E, Matthews FE, Wareham N, Brayne C, Khaw KT. Understanding the relationship between cognition and death: a within cohort examination of cognitive measures and mortality. Eur J Epidemiol 33: 1049–1062, 2018. doi:10.1007/s10654-018-0439-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 354. Batty GD, Deary IJ, Zaninotto P. Association of cognitive function with cause-specific mortality in middle and older age: follow-up of participants in the English Longitudinal Study of Ageing. Am J Epidemiol 183: 183–190, 2016. doi:10.1093/aje/kwv139.
    Crossref | PubMed | Web of Science | Google Scholar
  • 355. Wang Y, Du Y, Li J, Qiu C. lifespan intellectual factors, genetic susceptibility, and cognitive phenotypes in aging: implications for interventions. Front Aging Neurosci 11: 129, 2019. doi:10.3389/fnagi.2019.00129.
    Crossref | PubMed | Web of Science | Google Scholar
  • 356. Liu H, Fan Y, Liang J, Hu A, Chen W, Wang H, Fan Y, Li M, Duan J, Wang Q. A causal relationship between sarcopenia and cognitive impairment: a Mendelian randomization study. PLoS One 19: e0309124, 2024. doi:10.1371/journal.pone.0309124.
    Crossref | PubMed | Web of Science | Google Scholar
  • 357. Li S, Wang P, Cai Z, Jiang W, Xin X, Wang X, Zhou X. Correlates of physical activity levels, muscle strength, working memory, and cognitive function in older adults. Front Aging Neurosci 15: 1283864, 2023. doi:10.3389/fnagi.2023.1283864.
    Crossref | PubMed | Web of Science | Google Scholar
  • 358. Blackmore DG, Schaumberg MA, Ziaei M, Belford S, To XV, O’Keeffe I, Bernard A, Mitchell J, Hume E, Rose GL, Shaw T, York A, Barth M, Cooper EJ, Skinner TL, Nasrallah F, Riek S, Bartlett PF. Long-term improvement in hippocampal-dependent learning ability in healthy, aged individuals following high intensity interval training. Aging Dis, 2024. doi:10.14336/AD.2024.0642.
    Crossref | PubMed | Web of Science | Google Scholar
  • 359. Dupuy O, Ludyga S, Ortega FB, Hillman CH, Erickson KI, Herold F, Kamijo K, Wang CH, Morris TP, Brown B, Esteban-Cornejo I, Solis-Urra P, Bosquet L, Gerber M, Mekari S, Berryman N, Bherer L, Rattray B, Liu-Ambrose T, Voelcker-Rehage C, Cheval B. Do not underestimate the cognitive benefits of exercise. Nat Hum Behav 8: 1460–1463, 2024. doi:10.1038/s41562-024-01949-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 360. Ciria LF, Román-Caballero R, Vadillo MA, Holgado D, Luque-Casado A, Perakakis P, Sanabria D. Reply to: Do not underestimate the cognitive benefits of exercise. Nat Hum Behav 8: 1464–1466, 2024. doi:10.1038/s41562-024-01950-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 361. Gomez-Pinilla F, Hillman C. The influence of exercise on cognitive abilities. Compr Physiol 3: 403–428, 2013. doi:10.1002/cphy.c110063.
    Crossref | PubMed | Web of Science | Google Scholar
  • 362. Yu Y, Wang J, Xu J. Optimal dose and type of exercise to improve cognitive function in patients with mild cognitive impairment: a systematic review and network meta-analysis of RCTs. Front Psychiatry 15: 1436499, 2024. doi:10.3389/fpsyt.2024.1436499.
    Crossref | PubMed | Web of Science | Google Scholar
  • 363. Gallardo-Gómez D, Del Pozo-Cruz J, Noetel M, Álvarez-Barbosa F, Alfonso-Rosa RM, Del Pozo Cruz B. Optimal dose and type of exercise to improve cognitive function in older adults: A systematic review and bayesian model-based network meta-analysis of RCTs. Ageing Res Rev 76: 101591, 2022. doi:10.1016/j.arr.2022.101591.
    Crossref | PubMed | Web of Science | Google Scholar
  • 364. Zhang Y, DeFina LF, Leonard D, Chen B, Hébert ET, Barlow CE, Pavlovic A, Kohl HW 3rd. Associations of muscle-strengthening activity and cognitive function in community-dwelling middle-aged and older adults. J Aging Health 2024: 8982643241307757, 2024. doi:10.1177/08982643241307757.
    Crossref | PubMed | Web of Science | Google Scholar
  • 365. Reparaz-Escudero I, Izquierdo M, Bischoff-Ferrari HA, Martínez-Lage P, Saez de Asteasu ML. Effect of long-term physical exercise and multidomain interventions on cognitive function and the risk of mild cognitive impairment and dementia in older adults: a systematic review with meta-analysis. Ageing Res Rev 100: 102463, 2024. doi:10.1016/j.arr.2024.102463.
    Crossref | PubMed | Web of Science | Google Scholar
  • 366. Brodaty H, Chau T, Heffernan M, Ginige JA, Andrews G, Millard M, Sachdev PS, Anstey KJ, Lautenschlager NT, McNeil JJ, Jorm L, Kochan NA, Maeder A, Welberry H, San Jose JC, Briggs NE, Popovic G, Mavros Y, Almendrales Rangel C, Noble Y, Radd-Vagenas S, Flood VM, O’Leary F, Lampit A, Walton CC, Barr P, Fiatarone Singh M, Valenzuela M. An online multidomain lifestyle intervention to prevent cognitive decline in at-risk older adults: a randomized controlled trial. Nat Med 31: 565–573, 2025. doi:10.1038/s41591-024-03351-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 367. Wang W, Sun P, Lv T, Li M. The impact of modifiable health metrics on mortality for older adults with low cognitive function. Front Public Health 12: 1304876, 2024. doi:10.3389/fpubh.2024.1304876.
    Crossref | PubMed | Web of Science | Google Scholar
  • 368. Halma M, Marik P, Gazda S, Tuszynski J. Lifestyle medicine for healthy cognitive aging: a narrative review. Brain Behav Immun Integr 7: 100077, 2024. doi:10.1016/j.bbii.2024.100077.
    Crossref | PubMed | Google Scholar
  • 369. Tian Q, Greig EE, Davatzikos C, Landman BA, Resnick SM, Ferrucci L. Higher skeletal muscle mitochondrial oxidative capacity is associated with preserved brain structure up to over a decade. Nat Commun 15: 10786, 2024. doi:10.1038/s41467-024-55009-z.
    Crossref | Web of Science | Google Scholar
  • 370. Kelly NA, Wood KH, Allendorfer JB, Ford MP, Bickel CS, Marstrander J, Amara AW, Anthony T, Bamman MM, Skidmore FM. High-intensity exercise acutely increases substantia nigra and prefrontal brain activity in Parkinson’s disease. Med Sci Monit 23: 6064–6071, 2017. doi:10.12659/msm.906179.
    Crossref | PubMed | Web of Science | Google Scholar
  • 371. Zhan Y, Zhang Z, Lin S, Du B, Zhang K, Wu J, Xu H. Causal association of sarcopenia-related traits with brain cortical structure: a bidirectional Mendelian randomization study. Aging Clin Exp Res 37: 57, 2025. doi:10.1007/s40520-025-02977-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 372. Colcombe SJ, Kramer AF, Erickson KI, Scalf P, McAuley E, Cohen NJ, Webb A, Jerome GJ, Marquez DX, Elavsky S. Cardiovascular fitness, cortical plasticity, and aging. Proc Natl Acad Sci USA 101: 3316–3321, 2004. doi:10.1073/pnas.0400266101.
    Crossref | PubMed | Web of Science | Google Scholar
  • 373. Ferrer-Uris B, Ramos MA, Busquets A, Angulo-Barroso R. Can exercise shape your brain? A review of aerobic exercise effects on cognitive function and neuro-physiological underpinning mechanisms. AIMS Neurosci 9: 150–174, 2022. doi:10.3934/Neuroscience.2022009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 374. Thomas AG, Dennis A, Bandettini PA, Johansen-Berg H. The effects of aerobic activity on brain structure. Front Psychol 3: 86, 2012. doi:10.3389/fpsyg.2012.00086.
    Crossref | PubMed | Web of Science | Google Scholar
  • 375. Erickson KI, Raji CA, Lopez OL, Becker JT, Rosano C, Newman AB, Gach HM, Thompson PM, Ho AJ, Kuller LH. Physical activity predicts gray matter volume in late adulthood: the Cardiovascular Health Study. Neurology 75: 1415–1422, 2010. doi:10.1212/WNL.0b013e3181f88359.
    Crossref | PubMed | Web of Science | Google Scholar
  • 376. Colcombe SJ, Erickson KI, Scalf PE, Kim JS, Prakash R, McAuley E, Elavsky S, Marquez DX, Hu L, Kramer AF. Aerobic exercise training increases brain volume in aging humans. J Gerontol A Biol Sci Med Sci 61: 1166–1170, 2006. doi:10.1093/gerona/61.11.1166.
    Crossref | PubMed | Web of Science | Google Scholar
  • 377. Ferrer-Ramos P, Garnacho-Castaño MV, Girabent-Farrés M, Faundez-Zanuy M, Serra-Payá N. Physical performance tests for preliminary cognitive screening in older adults: a systematic review of strength, walking, and balance assessments. Arch Gerontol Geriatr 130: 105722, 2025. doi:10.1016/j.archger.2024.105722.
    Crossref | PubMed | Web of Science | Google Scholar
  • 378. Jia YJ, Wang J, Ren JR, Chan P, Chen SD, Chen XC, , et al.. A framework of biomarkers for brain aging: a consensus statement by the Aging Biomarker Consortium. Life Med 2: lnad017, 2023. doi:10.1093/lifemedi/lnad017.
    Crossref | PubMed | Web of Science | Google Scholar
  • 379. Walhovd KB, Lövden M, Fjell AM. Timing of lifespan influences on brain and cognition. Trends Cogn Sci 27: 901–915, 2023. doi:10.1016/j.tics.2023.07.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 380. McCracken C, Raisi-Estabragh Z, Szabo L, Robson J, Raman B, Topiwala A, Roca-Fernández A, Husain M, Petersen SE, Neubauer S, Nichols TE. NHS Health Check attendance is associated with reduced multiorgan disease risk: a matched cohort study in the UK Biobank. BMC Med 22: 1, 2024. doi:10.1186/s12916-023-03187-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 381. Krogsbøll LT, Jørgensen KJ, Gøtzsche PC. General health checks in adults for reducing morbidity and mortality from disease. Cochrane Database Syst Rev 1: CD009009, 2019. doi:10.1002/14651858.CD009009.pub3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 382. Shattock AJ, Johnson HC, Sim SY, Carter A, Lambach P, Hutubessy RC, Thompson KM, Badizadegan K, Lambert B, Ferrari MJ, Jit M, Fu H, Silal SP, Hounsell RA, White RG, Mosser JF, Gaythorpe KA, Trotter CL, Lindstrand A, O’Brien KL, Bar-Zeev N. Contribution of vaccination to improved survival and health: modelling 50 years of the Expanded Programme on Immunization. Lancet 403: 2307–2316, 2024. doi:10.1016/S0140-6736(24)00850-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 383. Mendoza-Cano O, Trujillo X, Huerta M, Rios-Silva M, Guzman-Esquivel J, Lugo-Radillo A, Benites-Godínez V, Bricio-Barrios JA, Cárdenas-Rojas MI, Ríos-Bracamontes EF, Guzman-Solorzano HP, Baltazar-Rodríguez GM, Ruiz-Montes de Oca V, Ortega-Macías VM, Ortega-Ramírez AD, Murillo-Zamora E. Assessing the influence of COVID-19 vaccination coverage on excess mortality across 178 countries: a cross-sectional study. Vaccines (Basel) 11, 2023. doi:10.3390/vaccines11081294.
    Crossref | PubMed | Web of Science | Google Scholar
  • 384. Chang KC, Lee JT, Vamos EP, Soljak M, Johnston D, Khunti K, Majeed A, Millett C. Impact of the National Health Service Health Check on cardiovascular disease risk: a difference-in-differences matching analysis. CMAJ 188: E228–E238, 2016. doi:10.1503/cmaj.151201.
    Crossref | PubMed | Web of Science | Google Scholar
  • 385. Debiec R, Lawday D, Bountziouka V, Beeston E, Greengrass C, Bramley R, Sehmi S, Kharodia S, Newton M, Marshall A, Krzeminski A, Zafar A, Chahal A, Heer A, Khunti K, Joshi N, Lakhani M, Farooqi A, Patel R, Samani NJ. Evaluating the clinical effectiveness of the NHS Health Check programme: a prospective analysis in the Genetics and Vascular Health Check (GENVASC) study. BMJ Open 13: e068025, 2023. doi:10.1136/bmjopen-2022-068025.
    Crossref | PubMed | Web of Science | Google Scholar
  • 386. Alageel S, Gulliford MC. Health checks and cardiovascular risk factor values over six years’ follow-up: matched cohort study using electronic health records in England. PLoS Med 16: e1002863, 2019. doi:10.1371/journal.pmed.1002863.
    Crossref | PubMed | Web of Science | Google Scholar
  • 387. Border R, Corley RP, Brown SA, Hewitt JK, Hopfer CJ, McWilliams SK, Rhea SA, Shriver CL, Stallings MC, Wall TL, Woodward KE, Rhee SH. Independent predictors of mortality in adolescents ascertained for conduct disorder and substance use problems, their siblings and community controls. Addiction 113: 2107–2115, 2018. doi:10.1111/add.14366.
    Crossref | PubMed | Web of Science | Google Scholar
  • 388. Tariq N, Gupta V. High risk behaviors. In: StatPearls. StatPearls Publishing, 2025.
    Google Scholar
  • 389. Woolf SH, Schoomaker H. Life expectancy and mortality rates in the United States, 1959-2017. JAMA 322: 1996–2016, 2019. doi:10.1001/jama.2019.16932.
    Crossref | PubMed | Web of Science | Google Scholar
  • 390. Sakran JV, Lunardi N. Reducing firearm injury and death in the United States. Adv Surg 56: 49–67, 2022. doi:10.1016/j.yasu.2022.03.001.
    Crossref | PubMed | Google Scholar
  • 391. Greenberg B, Bennett A, Naveed A, Petrut R, Wang SM, Vyas N, Bachari A, Khan S, Sue TC, Dryburgh N, Almoli F, Skidmore B, Shaver N, Bui EC, Brouwers M, Moher D, Little J, Maggi J, Ahmed N. How firearm legislation impacts firearm mortality internationally: a scoping review. Health Policy Open 7: 100127, 2024. doi:10.1016/j.hpopen.2024.100127.
    Crossref | PubMed | Web of Science | Google Scholar
  • 392. Naghavi M, Marczak LB, Kutz M, Shackelford KA, Arora M, Miller-Petrie M, , et al.. Global mortality from firearms, 1990-2016. JAMA 320: 792–814, 2018. doi:10.1001/jama.2018.10060.
    Crossref | PubMed | Web of Science | Google Scholar
  • 393. Goldstick JE, Cunningham RM, Carter PM. Current causes of death in children and adolescents in the United States. N Engl J Med 386: 1955–1956, 2022. doi:10.1056/NEJMc2201761.
    Crossref | PubMed | Web of Science | Google Scholar
  • 394. Roser M. Why is life expectancy in the US lower than in other rich countries? OurWorldinData.org. https://ourworldindata.org/us-life-expectancy-low [2024 Sep 23].
    Google Scholar
  • 395. GBD 2015 Tobacco Collaborators. Smoking prevalence and attributable disease burden in 195 countries and territories, 1990–2015: a systematic analysis from the Global Burden of Disease Study 2015. Lancet 389: 1885–1906, 2017. doi:10.1016/S0140-6736(17)30819-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 396. GBD 2021 Tobacco Forecasting Collaborators. Forecasting the effects of smoking prevalence scenarios on years of life lost and life expectancy from 2022 to 2050: a systematic analysis for the Global Burden of Disease Study 2021. Lancet Public Health 9: e729–e744, 2024. doi:10.1016/S2468-2667(24)00166-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 397. Wensink M, Alvarez JA, Rizzi S, Janssen F, Lindahl-Jacobsen R. Progression of the smoking epidemic in high-income regions and its effects on male-female survival differences: a cohort-by-age analysis of 17 countries. BMC Public Health 20: 39, 2020. doi:10.1186/s12889-020-8148-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 398. Janssen F. Changing contribution of smoking to the sex differences in life expectancy in Europe, 1950-2014. Eur J Epidemiol 35: 835–841, 2020. doi:10.1007/s10654-020-00602-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 399. Rossow I, Amundsen A. Alcohol abuse and mortality: a 40-year prospective study of Norwegian conscripts. Soc Sci Med 44: 261–267, 1997. doi:10.1016/s0277-9536(96)00161-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 400. Alpert HR, Slater ME, Yoon YH, Chen CM, Winstanley N, Esser MB. Alcohol consumption and 15 causes of fatal injuries: a systematic review and meta-analysis. Am J Prev Med 63: 286–300, 2022. doi:10.1016/j.amepre.2022.03.025.
    Crossref | PubMed | Web of Science | Google Scholar
  • 401. Kim AM. Alcohol consumption and suicide rate: a cross-sectional analysis in 183 countries. Psychiatry Res 295: 113553, 2021. doi:10.1016/j.psychres.2020.113553.
    Crossref | PubMed | Web of Science | Google Scholar
  • 402. Hébert AH, Hill AL. Impact of opioid overdoses on US life expectancy and years of life lost, by demographic group and stimulant co-involvement: a mortality data analysis from 2019 to 2022. Lancet Reg Health Am 36: 100813, 2024. doi:10.1016/j.lana.2024.100813.
    Crossref | PubMed | Web of Science | Google Scholar
  • 403. Tian F, Shen Q, Hu Y, Ye W, Valdimarsdóttir UA, Song H, Fang F. Association of stress-related disorders with subsequent risk of all-cause and cause-specific mortality: a population-based and sibling-controlled cohort study. Lancet Reg Health Eur 18: 100402, 2022. doi:10.1016/j.lanepe.2022.100402.
    Crossref | PubMed | Web of Science | Google Scholar
  • 404. Rutters F, Pilz S, Koopman AD, Rauh SP, Te Velde SJ, Stehouwer CD, Elders PJ, Nijpels G, Dekker JM. The association between psychosocial stress and mortality is mediated by lifestyle and chronic diseases: the Hoorn Study. Soc Sci Med 118: 166–172, 2014. doi:10.1016/j.socscimed.2014.08.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 405. Prior A, Fenger-Grøn M, Larsen KK, Larsen FB, Robinson KM, Nielsen MG, Christensen KS, Mercer SW, Vestergaard M. The association between perceived stress and mortality among people with multimorbidity: a prospective population-based cohort study. Am J Epidemiol 184: 199–210, 2016. doi:10.1093/aje/kwv324.
    Crossref | PubMed | Web of Science | Google Scholar
  • 406. Aldwin CM, Molitor NT, Avron S 3rd, Levenson MR, Molitor J, Igarashi H. Do Stress trajectories predict mortality in older men? Longitudinal findings from the VA Normative Aging Study. J Aging Res 2011: 896109, 2011. doi:10.4061/2011/896109.
    Crossref | PubMed | Google Scholar
  • 407. Yoon ES, So WY, Jang S. Association between perceived psychological stress and exercise behaviors: a cross-sectional study using the survey of national physical fitness. Life (Basel) 13: 2059, 2023. doi:10.3390/life13102059.
    Crossref | Web of Science | Google Scholar
  • 408. Koga HK, Trudel-Fitzgerald C, Lee LO, James P, Kroenke C, Garcia L, Shadyab AH, Salmoirago-Blotcher E, Manson JE, Grodstein F, Kubzansky LD. Optimism, lifestyle, and longevity in a racially diverse cohort of women. J Am Geriatr Soc 70: 2793–2804, 2022. doi:10.1111/jgs.17897.
    Crossref | PubMed | Web of Science | Google Scholar
  • 409. Lee LO, James P, Zevon ES, Kim ES, Trudel-Fitzgerald C, Spiro A 3rd, Grodstein F, Kubzansky LD. Optimism is associated with exceptional longevity in 2 epidemiologic cohorts of men and women. Proc Natl Acad Sci USA 116: 18357–18362, 2019. doi:10.1073/pnas.1900712116.
    Crossref | PubMed | Web of Science | Google Scholar
  • 410. Rozanski A, Bavishi C, Kubzansky LD, Cohen R. Association of optimism with cardiovascular events and all-cause mortality: a systematic review and meta-analysis. JAMA Netw Open 2: e1912200, 2019. doi:10.1001/jamanetworkopen.2019.12200.
    Crossref | PubMed | Web of Science | Google Scholar
  • 411. Ren J, Mao C. Social relationships and mortality. Nat Hum Behav 7: 1249–1250, 2023. doi:10.1038/s41562-023-01611-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 412. Holt-Lunstad J. Social connection as a public health issue: the evidence and a systemic framework for prioritizing the “social” in social determinants of health. Annu Rev Public Health 43: 193–213, 2022. doi:10.1146/annurev-publhealth-052020-110732.
    Crossref | PubMed | Web of Science | Google Scholar
  • 413. Poulin MJ, Brown SL, Dillard AJ, Smith DM. Giving to others and the association between stress and mortality. Am J Public Health 103: 1649–1655, 2013. doi:10.2105/AJPH.2012.300876.
    Crossref | PubMed | Web of Science | Google Scholar
  • 414. Hawkley LC. Loneliness and health. Nat Rev Dis Primers 8: 22, 2022. doi:10.1038/s41572-022-00355-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 415. Sidik SM. Why loneliness is bad for your health. Nature 628: 22–24, 2024. doi:10.1038/d41586-024-00900-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 416. Ye L, Bally E, Korenhof SA, Fierloos I, Alhambra Borrás T, Clough G, Raat H, van Grieken A. The association between loneliness and frailty among community-dwelling older adults in five European countries: a longitudinal study. Age Ageing 53: afae210, 2024. doi:10.1093/ageing/afae210.
    Crossref | PubMed | Web of Science | Google Scholar
  • 417. Lin HY, Lin YC, Chen LK, Hsiao FY. Untangling the complex interplay between social isolation, anorexia, sarcopenia, and mortality: insights from a longitudinal study. J Nutr Health Aging 27: 797–805, 2023. doi:10.1007/s12603-023-1993-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 418. Liang YY, Zhou M, He Y, Zhang W, Wu Q, Luo T, Zhang J, Jia F, Qi L, Ai S, Zhang J. Observational and genetic evidence disagree on the association between loneliness and risk of multiple diseases. Nat Hum Behav 8: 2209–2221, 2024. doi:10.1038/s41562-024-01970-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 419. Alfredsson L, Armstrong BK, Butterfield DA, Chowdhury R, de Gruijl FR, Feelisch M, Garland CF, Hart PH, Hoel DG, Jacobsen R, Lindqvist PG, Llewellyn DJ, Tiemeier H, Weller RB, Young AR. Insufficient sun exposure has become a real public health problem. Int J Environ Res Public Health 17: 5014, 2020. doi:10.3390/ijerph17145014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 420. Weller RB. Sunlight: time for a rethink? J Invest Dermatol 144: 1724–1732, 2024. doi:10.1016/j.jid.2023.12.027.
    Crossref | PubMed | Web of Science | Google Scholar
  • 421. Stevenson AC, Clemens T, Pairo-Castineira E, Webb DJ, Weller RB, Dibben C. Higher ultraviolet light exposure is associated with lower mortality: an analysis of data from the UK biobank cohort study. Health Place 89: 103328, 2024. doi:10.1016/j.healthplace.2024.103328.
    Crossref | PubMed | Web of Science | Google Scholar
  • 422. Breakell T, Kowalski I, Foerster Y, Kramer R, Erdmann M, Berking C, Heppt MV. Ultraviolet filters: dissecting current facts and myths. J Clin Med 13: 2986, 2024. doi:10.3390/jcm13102986.
    Crossref | PubMed | Web of Science | Google Scholar
  • 423. Sander M, Sander M, Burbidge T, Beecker J. The efficacy and safety of sunscreen use for the prevention of skin cancer. CMAJ 192: E1802–E1808, 2020. doi:10.1503/cmaj.201085.
    Crossref | PubMed | Web of Science | Google Scholar
  • 424. Global Burden of Disease US Health Disparities Collaborators. Life expectancy by county and educational attainment in the USA, 2000-19: an observational analysis. Lancet Public Health 10: E136–E147, 2025. doi:10.1016/S2468-2667(24)00303-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 425. Stringhini S, Carmeli C, Jokela M, Avendaño M, Muennig P, Guida F, , et al.. Socioeconomic status and the 25 x 25 risk factors as determinants of premature mortality: a multicohort study and meta-analysis of 1.7 million men and women. Lancet 389: 1229–1237, 2017. doi:10.1016/S0140-6736(16)32380-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 426. Mackenbach JP, Stirbu I, Roskam AJ, Schaap MM, Menvielle G, Leinsalu M, Kunst AE; European Union Working Group on Socioeconomic Inequalities in Health. Socioeconomic inequalities in health in 22 European countries. N Engl J Med 358: 2468–2481, 2008. doi:10.1056/NEJMsa0707519.
    Crossref | PubMed | Web of Science | Google Scholar
  • 427. Hajat A, Kaufman JS, Rose KM, Siddiqi A, Thomas JC. Long-term effects of wealth on mortality and self-rated health status. Am J Epidemiol 173: 192–200, 2011. doi:10.1093/aje/kwq348.
    Crossref | PubMed | Web of Science | Google Scholar
  • 428. Link BG, Phelan J. Social conditions as fundamental causes of disease. J Health Soc Behav, 35: 80–94, 1995. doi:10.2307/2626958.
    Crossref | Web of Science | Google Scholar
  • 429. Chetty R, Stepner M, Abraham S, Lin S, Scuderi B, Turner N, Bergeron A, Cutler D. The association between income and life expectancy in the United States, 2001-2014. JAMA 315: 1750–1766, 2016. doi:10.1001/jama.2016.4226.
    Crossref | PubMed | Web of Science | Google Scholar
  • 430. Marmot M. The health gap: the challenge of an unequal world: the argument. Int J Epidemiol 46: 1312–1318, 2017. doi:10.1093/ije/dyx163.
    Crossref | PubMed | Web of Science | Google Scholar
  • 431. Magnan S. Social determinants of health 101 for health care: five plus five. NAM Perspect 7, 2017. doi:10.31478/201710c.
    Crossref | Google Scholar
  • 432. Hood CM, Gennuso KP, Swain GR, Catlin BB. County health rankings: relationships between determinant factors and health outcomes. Am J Prev Med 50: 129–135, 2016. doi:10.1016/j.amepre.2015.08.024.
    Crossref | PubMed | Web of Science | Google Scholar
  • 433. Stringhini S, Carmeli C, Jokela M, Avendaño M, McCrory C, d’Errico A, Bochud M, Barros H, Costa G, Chadeau-Hyam M, Delpierre C, Gandini M, Fraga S, Goldberg M, Giles GG, Lassale C, Kenny RA, Kelly-Irving M, Paccaud F, Layte R, Muennig P, Marmot MG, Ribeiro AI, Severi G, Steptoe A, Shipley MJ, Zins M, Mackenbach JP, Vineis P, Kivimäki M; LIFEPATH Consortium. Socioeconomic status, non-communicable disease risk factors, and walking speed in older adults: multi-cohort population based study. BMJ 360: k1046, 2018. doi:10.1136/bmj.k1046.
    Crossref | PubMed | Web of Science | Google Scholar
  • 434. Turrell G, Lynch JW, Leite C, Raghunathan T, Kaplan GA. Socioeconomic disadvantage in childhood and across the life course and all-cause mortality and physical function in adulthood: evidence from the Alameda County Study. J Epidemiol Community Health 61: 723–730, 2007. doi:10.1136/jech.2006.050609.
    Crossref | PubMed | Web of Science | Google Scholar
  • 435. Belsky DW, Caspi A, Arseneault L, Corcoran DL, Domingue BW, Harris KM, Houts RM, Mill JS, Moffitt TE, Prinz J, Sugden K, Wertz J, Williams B, Odgers CL. Genetics and the geography of health, behaviour and attainment. Nat Hum Behav 3: 576–586, 2019. doi:10.1038/s41562-019-0562-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 436. Sud A, Horton RH, Hingorani AD, Tzoulaki I, Turnbull C, Houlston RS, Lucassen A. Realistic expectations are key to realising the benefits of polygenic scores. BMJ 380: e073149, 2023. doi:10.1136/bmj-2022-073149.
    Crossref | PubMed | Web of Science | Google Scholar
  • 437. McLoughlin GM, Salmon J. How can we equitably scale-up physical activity interventions to ensure everyone has opportunities to thrive? J Phys Act Health 21: 729–730, 2024. doi:10.1123/jpah.2024-0342.
    Crossref | PubMed | Web of Science | Google Scholar
  • 438. Harris D, Dlima SD, Gluchowski A, Hall A, Elliott E, Munford L. The effectiveness and acceptability of physical activity interventions amongst older adults with lower socioeconomic status: a mixed methods systematic review. Int J Behav Nutr Phys Act 21: 121, 2024. doi:10.1186/s12966-024-01666-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 439. Walensky RP, McCann NC. Challenges to the future of a robust physician workforce in the United States. N Engl J Med 392: 286–295, 2025. doi:10.1056/NEJMsr2412784.
    Crossref | PubMed | Web of Science | Google Scholar
  • 440. Dieleman JL, Weil M, Beauchamp M, Bisignano C, Crosby SW, DeJarnatt D, Lescinsky H, Mokdad AH, Ostroff S, Paul H, Pollock I, Sahu M, Scott JW, Taylor KV, Thomson A, Weaver MR, Wilner LB, Murray CJ. Drivers of variation in health care spending across US counties. JAMA Health Forum 6: e245220, 2025. doi:10.1001/jamahealthforum.2024.5220.
    Crossref | Web of Science | Google Scholar
  • 441. Kruk ME, Gage AD, Joseph NT, Danaei G, García-Saisó S, Salomon JA. Mortality due to low-quality health systems in the universal health coverage era: a systematic analysis of amenable deaths in 137 countries. Lancet 392: 2203–2212, 2018. doi:10.1016/S0140-6736(18)31668-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 442. GBD 2015 Healthcare Access and Quality Collaborators. Healthcare Access and Quality Index based on mortality from causes amenable to personal health care in 195 countries and territories, 1990–2015: a novel analysis from the Global Burden of Disease Study 2015. Lancet 390: 231–266, 2017. doi:10.1016/S0140-6736(17)30818-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 443. Global Nutrition Target Collaborators. Global, regional, and national progress towards the 2030 global nutrition targets and forecasts to 2050: a systematic analysis for the Global Burden of Disease Study 2021. Lancet 404: 2543–2583, 2025. doi:10.1016/S0140-6736(24)01821-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 444. Gunja MZ, Gumas ED, Williams RD 2nd. U.S. Health Care from a Global Perspective, 2022: Accelerating Spending, Worsening Outcomes. Commonwealth Fund, 2022. https://www.commonwealthfund.org/publications/issue-briefs/2023/jan/us-health-care-global-perspective-2022.
    Google Scholar
  • 445. Papanicolas I, Woskie LR, Jha AK. Health care spending in the United States and other high-income countries. JAMA 319: 1024–1039, 2018. doi:10.1001/jama.2018.1150.
    Crossref | PubMed | Web of Science | Google Scholar
  • 446. Lorenzoni L, Belloni A, Sassi F. Health-care expenditure and health policy in the USA versus other high-spending OECD countries. Lancet 384: 83–92, 2014. doi:10.1016/S0140-6736(14)60571-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 447. Blumenthal D, Gumas E, Shah A. The failing U.S. health system. N Engl J Med 391: 1566–1568, 2024. doi:10.1056/NEJMp2410855.
    Crossref | PubMed | Web of Science | Google Scholar
  • 448. GBD 2021 US Burden of Disease and Forecasting Collaborators. Burden of disease scenarios by state in the USA, 2022-50: a forecasting analysis for the Global Burden of Disease Study 2021. Lancet 404: 2341–2370, 2024. doi:10.1016/S0140-6736(24)02246-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 449. GBD 2021 US Burden of Disease Collaborators. The burden of diseases, injuries, and risk factors by state in the USA, 1990-2021: a systematic analysis for the Global Burden of Disease Study 2021. Lancet 404: 2314–2340, 2024. doi:10.1016/S0140-6736(24)01446-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 450. Hoyert D. Maternal Mortality Rates in the United States, 2021. NCHS Health E-Stats, 2023. https://www.cdc.gov/nchs/data/hestat/maternal-mortality/2023/maternal-mortality-rates-2023.htm.
    Crossref | Google Scholar
  • 451. Murphy SL, Kochanek KD, Xu J, Arias E. Mortality in the United States, 2023. NCHS Data Brief 521: CS356116, 2024. doi:10.15620/cdc/170564.
    Crossref | Google Scholar
  • 452. Woolf SH. Understanding disparities in life expectancy. Lancet 404: 2243–2244, 2024. doi:10.1016/S0140-6736(24)02136-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 453. Dwyer-Lindgren L, Baumann MM, Li Z, Kelly YO, Schmidt C, Searchinger C, La Motte-Kerr W, Bollyky TJ, Mokdad AH, Murray CJ. Ten Americas: a systematic analysis of life expectancy disparities in the USA. Lancet 404: 2299–2313, 2024. doi:10.1016/S0140-6736(24)01495-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 454. The Lancet. Health in the age of disinformation. Lancet 405: 173, 2025. doi:10.1016/S0140-6736(25)00094-7.
    Crossref | Web of Science | Google Scholar
  • 455. Niederdeppe J, Boyd AD, King AJ, Rimal RN. Strategies for effective public health communication in a complex information environment. Annu Rev Public Health 46: 411–431, 2025. doi:10.1146/annurev-publhealth-071723-120721.
    Crossref | PubMed | Web of Science | Google Scholar
  • 456. Calleja-Agius J, England K, Calleja N. The effect of global warming on mortality. Early Hum Dev 155: 105222, 2021. doi:10.1016/j.earlhumdev.2020.105222.
    Crossref | PubMed | Web of Science | Google Scholar
  • 457. Luthi S, Fairless C, Fischer EM, Scovronick N, Ben A, Coelho M, Guo YL, Guo Y, Honda Y, Huber V, Kysely J, Lavigne E, Roye D, Ryti N, Silva S, Urban A, Gasparrini A, Bresch DN, Vicedo-Cabrera AM. Rapid increase in the risk of heat-related mortality. Nat Commun 14: 4894, 2023. doi:10.1038/s41467-023-40599-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 458. Howard JT, Androne N, Alcover KC, Santos-Lozada AR. Trends of heat-related deaths in the US, 1999–2023. JAMA 332: 1203–1204, 2024. doi:10.1001/jama.2024.16386.
    Crossref | PubMed | Web of Science | Google Scholar
  • 459. Romanello M, Walawender M, Hsu SC, Moskeland A, Palmeiro-Silva Y, Scamman D, , et al.. The 2024 report of the Lancet Countdown on health and climate change: facing record-breaking threats from delayed action. Lancet 404: 1847–1896, 2024. doi:10.1016/S0140-6736(24)01822-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 460. Liu C, Chen R, Sera F, Vicedo-Cabrera AM, Guo Y, Tong S, , et al.. Ambient particulate air pollution and daily mortality in 652 cities. N Engl J Med 381: 705–715, 2019. doi:10.1056/NEJMoa1817364.
    Crossref | PubMed | Web of Science | Google Scholar
  • 461. Jian Y, Messer LC, Jagai JS, Rappazzo KM, Gray CL, Grabich SC, Lobdell DT. Associations between environmental quality and mortality in the contiguous United States, 2000–2005. Environ Health Perspect 125: 355–362, 2017. doi:10.1289/EHP119.
    Crossref | PubMed | Web of Science | Google Scholar
  • 462. Drew L. Air pollution and brain damage: what the science says. Nature 637: 536–538, 2025. doi:10.1038/d41586-025-00053-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 463. Lee W, Choi HM, Kim D, Honda Y, Leon Guo YL, Kim H. Synergic effect between high temperature and air pollution on mortality in Northeast Asia. Environ Res 178: 108735, 2019. doi:10.1016/j.envres.2019.108735.
    Crossref | PubMed | Web of Science | Google Scholar
  • 464. Kozlov M. Landmark study links microplastics to serious health problems. Nature, 2024. doi:10.1038/d41586-024-00650-3.
    Crossref | PubMed | Google Scholar
  • 465. Marfella R, Prattichizzo F, Sardu C, Fulgenzi G, Graciotti L, Spadoni T, , et al.. Microplastics and nanoplastics in atheromas and cardiovascular events. N Engl J Med 390: 900–910, 2024. doi:10.1056/NEJMoa2309822.
    Crossref | PubMed | Web of Science | Google Scholar
  • 466. Landrigan PJ, Raps H, Cropper M, Bald C, Brunner M, Canonizado EM, , et al.. The Minderoo-Monaco commission on plastics and human health. Ann Glob Health 89: 23, 2023. doi:10.5334/aogh.4056.
    Crossref | PubMed | Web of Science | Google Scholar
  • 467. Thompson RC, Courtene-Jones W, Boucher J, Pahl S, Raubenheimer K, Koelmans AA. Twenty years of microplastic pollution research—what have we learned? Science 386: eadl2746, 2024. doi:10.1126/science.adl2746.
    Crossref | PubMed | Web of Science | Google Scholar
  • 468. Kozlov M. Your brain is full of microplastics: are they harming you? Nature 638: 311–313, 2025. doi:10.1038/d41586-025-00405-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 469. Fan Y, Tao C, Li Z, Huang Y, Yan W, Zhao S, Gao B, Xu Q, Qin Y, Wang X, Peng Z, Covaci A, Li Y, Xia Y, Lu C. Association of endocrine-disrupting chemicals with all-cause and cause-specific mortality in the U.S.: a prospective cohort study. Environ Sci Technol 57: 2877–2886, 2023. doi:10.1021/acs.est.2c07611.
    Crossref | PubMed | Web of Science | Google Scholar
  • 470. Cropper M, Dunlop S, Hinshaw H, Landrigan P, Park Y, Symeonides C. The benefits of removing toxic chemicals from plastics. Proc Natl Acad Sci USA 121: e2412714121, 2024. doi:10.1073/pnas.2412714121.
    Crossref | PubMed | Web of Science | Google Scholar
  • 471. Thacher JD, Roswall N, Ögren M, Pyko A, Åkesson A, Oudin A, , et al.. Residential exposure to transportation noise and risk of incident atrial fibrillation: a pooled study of 11 prospective Nordic cohorts. Lancet Reg Health Eur 46: 101091, 2024. doi:10.1016/j.lanepe.2024.101091.
    Crossref | PubMed | Web of Science | Google Scholar
  • 472. Sørensen M, Pershagen G, Thacher JD, Lanki T, Wicki B, Röösli M, Vienneau D, Cantuaria ML, Schmidt JH, Aasvang GM, Al-Kindi S, Osborne MT, Wenzel P, Sastre J, Fleming I, Schulz R, Hahad O, Kuntic M, Zielonka J, Sies H, Grune T, Frenis K, Münzel T, Daiber A. Health position paper and redox perspectives—disease burden by transportation noise. Redox Biol 69: 102995, 2024. doi:10.1016/j.redox.2023.102995.
    Crossref | PubMed | Web of Science | Google Scholar
  • 473. Topriceanu CC, Gong X, Shah M, Shiwani H, Eminson K, Atilola GO, Jephcote C, Adams K, Blangiardo M, Moon JC, Hughes AD, Gulliver J, Rowlands AV, Chaturvedi N, O’Regan DP, Hansell AL, Captur G. Higher aircraft noise exposure is linked to worse heart structure and function by cardiovascular MRI. J Am Coll Cardiol 85: 454–469, 2025. doi:10.1016/j.jacc.2024.09.1217.
    Crossref | PubMed | Web of Science | Google Scholar
  • 474. Panchin AY, Ogmen A, Blagodatski AS, Egorova A, Batin M, Glinin T. Targeting multiple hallmarks of mammalian aging with combinations of interventions. Aging (Albany NY) 16: 12073–12100, 2024. doi:10.18632/aging.206078.
    Crossref | PubMed | Google Scholar
  • 475. Handschin C. Caloric restriction and exercise “mimetics”: ready for prime time? Pharmacol Res 103: 158–166, 2016. doi:10.1016/j.phrs.2015.11.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 476. Weihrauch M, Handschin C. Pharmacological targeting of exercise adaptations in skeletal muscle: Benefits and pitfalls. Biochem Pharmacol 147: 211–220, 2018. doi:10.1016/j.bcp.2017.10.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 477. Gliemann L, Schmidt JF, Olesen J, Biensø RS, Peronard SL, Grandjean SU, Mortensen SP, Nyberg M, Bangsbo J, Pilegaard H, Hellsten Y. Resveratrol blunts the positive effects of exercise training on cardiovascular health in aged men. J Physiol 591: 5047–5059, 2013. doi:10.1113/jphysiol.2013.258061.
    Crossref | PubMed | Web of Science | Google Scholar
  • 478. Scribbans TD, Ma JK, Edgett BA, Vorobej KA, Mitchell AS, Zelt JG, Simpson CA, Quadrilatero J, Gurd BJ. Resveratrol supplementation does not augment performance adaptations or fibre-type-specific responses to high-intensity interval training in humans. Appl Physiol Nutr Metab 39: 1305–1313, 2014. doi:10.1139/apnm-2014-0070.
    Crossref | PubMed | Web of Science | Google Scholar
  • 479. Martinez-Negrin G, Acton JP, Cocksedge SP, Bailey SJ, Clifford T. The effect of dietary (poly)phenols on exercise-induced physiological adaptations: a systematic review and meta-analysis of human intervention trials. Crit Rev Food Sci Nutr 62: 2872–2887, 2022. doi:10.1080/10408398.2020.1860898.
    Crossref | PubMed | Web of Science | Google Scholar
  • 480. Konopka AR, Laurin JL, Schoenberg HM, Reid JJ, Castor WM, Wolff CA, Musci RV, Safairad OD, Linden MA, Biela LM, Bailey SM, Hamilton KL, Miller BF. Metformin inhibits mitochondrial adaptations to aerobic exercise training in older adults. Aging Cell 18: e12880, 2019. doi:10.1111/acel.12880.
    Crossref | PubMed | Web of Science | Google Scholar
  • 481. Moreno-Cabañas A, Morales-Palomo F, Alvarez-Jimenez L, Ortega JF, Mora-Rodriguez R. Effects of chronic metformin treatment on training adaptations in men and women with hyperglycemia: a prospective study. Obesity (Silver Spring) 30: 1219–1230, 2022. doi:10.1002/oby.23410.
    Crossref | PubMed | Web of Science | Google Scholar
  • 482. Kulkarni AS, Peck BD, Walton RG, Kern PA, Mar JC, Windham ST, Bamman MM, Barzilai N, Peterson CA. Metformin alters skeletal muscle transcriptome adaptations to resistance training in older adults. Aging (Albany NY) 12: 19852–19866, 2020. doi:10.18632/aging.104096.
    Crossref | PubMed | Web of Science | Google Scholar
  • 483. Walton RG, Dungan CM, Long DE, Tuggle SC, Kosmac K, Peck BD, Bush HM, Villasante Tezanos AG, McGwin G, Windham ST, Ovalle F, Bamman MM, Kern PA, Peterson CA. Metformin blunts muscle hypertrophy in response to progressive resistance exercise training in older adults: A randomized, double-blind, placebo-controlled, multicenter trial: the MASTERS trial. Aging Cell 18: e13039, 2019. doi:10.1111/acel.13039.
    Crossref | PubMed | Web of Science | Google Scholar
  • 484. Pilmark NS, Petersen-Bønding C, Holm NF, Johansen MY, Pedersen BK, Hansen KB, Karstoft K. The effect of metformin on self-selected exercise intensity in healthy, lean males: a randomized, crossover, counterbalanced trial. Front Endocrinol (Lausanne) 12: 599164, 2021. doi:10.3389/fendo.2021.599164.
    Crossref | PubMed | Web of Science | Google Scholar
  • 485. Bjelakovic G, Nikolova D, Gluud C. Antioxidant supplements and mortality. Curr Opin Clin Nutr Metab Care 17: 40–44, 2014. doi:10.1097/MCO.0000000000000009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 486. Ristow M, Zarse K, Oberbach A, Klöting N, Birringer M, Kiehntopf M, Stumvoll M, Kahn CR, Blüher M. Antioxidants prevent health-promoting effects of physical exercise in humans. Proc Natl Acad Sci USA 106: 8665–8670, 2009. doi:10.1073/pnas.0903485106.
    Crossref | PubMed | Web of Science | Google Scholar
  • 487. Teixeira VH, Valente HF, Casal SI, Marques AF, Moreira PA. Antioxidants do not prevent postexercise peroxidation and may delay muscle recovery. Med Sci Sports Exerc 41: 1752–1760, 2009. doi:10.1249/MSS.0b013e31819fe8e3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 488. Dutra MT, Alex S, Mota MR, Sales NB, Brown LE, Bottaro M. Effect of strength training combined with antioxidant supplementation on muscular performance. Appl Physiol Nutr Metab 43: 775–781, 2018. doi:10.1139/apnm-2017-0866.
    Crossref | PubMed | Web of Science | Google Scholar
  • 489. Higgins MR, Izadi A, Kaviani M. Antioxidants and exercise performance: with a focus on vitamin E and C supplementation. Int J Environ Res Public Health 17: 8452, 2020. doi:10.3390/ijerph17228452.
    Crossref | PubMed | Web of Science | Google Scholar
  • 490. Yu J, Laybutt DR, Kim LJ, Quek LE, Wu LE, Morris MJ, Youngson NA. Exercise-induced benefits on glucose handling in a model of diet-induced obesity are reduced by concurrent nicotinamide mononucleotide. Am J Physiol Endocrinol Metab 321: E176–E189, 2021. doi:10.1152/ajpendo.00446.2020.
    Link | Web of Science | Google Scholar
  • 491. Nadeeshani H, Li J, Ying T, Zhang B, Lu J. Nicotinamide mononucleotide (NMN) as an anti-aging health product—promises and safety concerns. J Adv Res 37: 267–278, 2022. doi:10.1016/j.jare.2021.08.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 492. Palmer RD, Vaccarezza M. Nicotinamide adenine dinucleotide and the sirtuins caution: pro-cancer functions. Aging Med (Milton) 4: 337–344, 2021. doi:10.1002/agm2.12184.
    Crossref | PubMed | Web of Science | Google Scholar
  • 493. Saleh TA, Whitson JA, Keiser P, Prasad P, Jenkins BC, Sodeinde T, Mann C, Rabinovitch PS, McReynolds MR, Sweetwyne MT. Metabolite accumulation from oral NMN supplementation drives aging-specific kidney inflammation (Preprint). bioRxiv 2024.04.09.588624, 2024. doi:10.1101/2024.04.09.588624.
    Google Scholar
  • 494. Bodine SC. The role of mTORC1 in the regulation of skeletal muscle mass. Fac Rev 11: 32, 2022. doi:10.12703/r/11-32.
    Crossref | PubMed | Google Scholar
  • 495. Lim C, Nunes EA, Currier BS, McLeod JC, Thomas AC, Phillips SM. An evidence-based narrative review of mechanisms of resistance exercise-induced human skeletal muscle hypertrophy. Med Sci Sports Exerc 54: 1546–1559, 2022. doi:10.1249/MSS.0000000000002929.
    Crossref | PubMed | Web of Science | Google Scholar
  • 496. Ogasawara R, Jensen TE, Goodman CA, Hornberger TA. Resistance exercise-induced hypertrophy: a potential role for rapamycin-insensitive mTOR. Exerc Sport Sci Rev 47: 188–194, 2019. doi:10.1249/JES.0000000000000189.
    Crossref | PubMed | Web of Science | Google Scholar
  • 497. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos GD. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3: 1014–1019, 2001. doi:10.1038/ncb1101-1014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 498. Ogasawara R, Fujita S, Hornberger TA, Kitaoka Y, Makanae Y, Nakazato K, Naokata I. The role of mTOR signalling in the regulation of skeletal muscle mass in a rodent model of resistance exercise. Sci Rep 6: 31142, 2016. doi:10.1038/srep31142.
    Crossref | PubMed | Web of Science | Google Scholar
  • 499. Drummond MJ, Fry CS, Glynn EL, Dreyer HC, Dhanani S, Timmerman KL, Volpi E, Rasmussen BB. Rapamycin administration in humans blocks the contraction-induced increase in skeletal muscle protein synthesis. J Physiol 587: 1535–1546, 2009. doi:10.1113/jphysiol.2008.163816.
    Crossref | PubMed | Web of Science | Google Scholar
  • 500. Furrer R, Handschin C. Molecular aspects of the exercise response and training adaptation in skeletal muscle. Free Radic Biol Med 223: 53–68, 2024. doi:10.1016/j.freeradbiomed.2024.07.026.
    Crossref | PubMed | Web of Science | Google Scholar
  • 501. Hawley JA, Joyner MJ, Green DJ. Mimicking exercise: what matters most and where to next? J Physiol 599: 791–802, 2021. doi:10.1113/JP278761.
    Crossref | PubMed | Web of Science | Google Scholar
  • 502. Hawley JA, Holloszy JO. Exercise: it’s the real thing! Nutr Rev 67: 172–178, 2009. doi:10.1111/j.1753-4887.2009.00185.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 503. Davies RW, Lynch AE, Kumar U, Jakeman PM. Characterisation of the muscle protein synthetic response to resistance exercise in healthy adults: a systematic review and exploratory meta-analysis. Transl Sports Med 2024: 3184356, 2024. doi:10.1155/2024/3184356.
    Crossref | PubMed | Web of Science | Google Scholar
  • 504. Trommelen J, van Lieshout GA, Nyakayiru J, Holwerda AM, Smeets JS, Hendriks FK, van Kranenburg JM, Zorenc AH, Senden JM, Goessens JP, Gijsen AP, van Loon LJ. The anabolic response to protein ingestion during recovery from exercise has no upper limit in magnitude and duration in vivo in humans. Cell Rep Med 4: 101324, 2023. doi:10.1016/j.xcrm.2023.101324.
    Crossref | PubMed | Web of Science | Google Scholar
  • 505. Gems D. The aging-disease false dichotomy: understanding senescence as pathology. Front Genet 6: 212, 2015. doi:10.3389/fgene.2015.00212.
    Crossref | PubMed | Web of Science | Google Scholar
  • 506. Moqri M, Herzog C, Poganik JR, Justice J, Belsky DW, Higgins-Chen A, Moskalev A, Fuellen G, Cohen AA, Bautmans I, Widschwendter M, Ding J, Fleming A, Mannick J, Han JJ, Zhavoronkov A, Barzilai N, Kaeberlein M, Cummings S, Kennedy BK, Ferrucci L, Horvath S, Verdin E, Maier AB, Snyder MP, Sebastiano V, Gladyshev VN; Biomarkers of Aging Consortium. Biomarkers of aging for the identification and evaluation of longevity interventions. Cell 186: 3758–3775, 2023. doi:10.1016/j.cell.2023.08.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 507. Jylhävä J, Pedersen NL, Hägg S. Biological age predictors. EBioMedicine 21: 29–36, 2017. doi:10.1016/j.ebiom.2017.03.046.
    Crossref | PubMed | Web of Science | Google Scholar
  • 508. Rutledge J, Oh H, Wyss-Coray T. Measuring biological age using omics data. Nat Rev Genet 23: 715–727, 2022. doi:10.1038/s41576-022-00511-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 509. Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Biomarkers of cellular senescence and aging: current state-of-the-art, challenges and future perspectives. Adv Biol (Weinh) 8: e2400079, 2024. doi:10.1002/adbi.202400079.
    Crossref | PubMed | Web of Science | Google Scholar
  • 510. Chen R, Wang Y, Zhang S, Bulloch G, Zhang J, Liao H, Shang X, Clark M, Peng Q, Ge Z, Cheng CY, Gao Y, He M, Zhu Z. Biomarkers of ageing: current state‐of‐art, challenges, and opportunities. MedComm Futur Med 2: e50, 2023. doi:10.1002/mef2.50.
    Crossref | Google Scholar
  • 511. Han JJ. The ticking of aging clocks. Trends Endocrinol Metab 35: 11–22, 2024. doi:10.1016/j.tem.2023.09.007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 512. Palmer RD. Aging clocks & mortality timers, methylation, glycomic, telomeric and more. A window to measuring biological age. Aging Med (Milton) 5: 120–125, 2022. doi:10.1002/agm2.12197.
    Crossref | PubMed | Web of Science | Google Scholar
  • 513. Tao X, Zhu Z, Wang L, Li C, Sun L, Wang W, Gong W. Biomarkers of aging and relevant evaluation techniques: a comprehensive review. Aging Dis 15: 977–1005, 2024. doi:10.14336/AD.2023.00808-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 514. Wagner KH, Cameron-Smith D, Wessner B, Franzke B. Biomarkers of aging: from function to molecular biology. Nutrients 8: 338, 2016. doi:10.3390/nu8060338.
    Crossref | PubMed | Web of Science | Google Scholar
  • 515. Ferrucci L, Gonzalez-Freire M, Fabbri E, Simonsick E, Tanaka T, Moore Z, Salimi S, Sierra F, de Cabo R. Measuring biological aging in humans: a quest. Aging Cell 19: e13080, 2020. doi:10.1111/acel.13080.
    Crossref | PubMed | Web of Science | Google Scholar
  • 516. de Lima Camillo LP, Asif MH, Horvath S, Larschan E, Singh R. Histone mark age of human tissues and cell types. Sci Adv 11: eadk9373, 2025. doi:10.1126/sciadv.adk9373.
    Crossref | PubMed | Web of Science | Google Scholar
  • 517. Oh HS, Rutledge J, Nachun D, Pálovics R, Abiose O, Moran-Losada P, , et al.. Organ aging signatures in the plasma proteome track health and disease. Nature 624: 164–172, 2023. doi:10.1038/s41586-023-06802-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 518. Goeminne LJ, Eames A, Tyshkovskiy A, Argentieri MA, Ying K, Moqri M, Gladyshev VN. Plasma-based organ-specific aging and mortality models unveil diseases as accelerated aging of organismal systems (Preprint). medRxiv 2024.04.08.24305469, 2024. doi:10.1101/2024.04.08.24305469.
    Google Scholar
  • 519. Carrasco-Zanini J, Pietzner M, Davitte J, Surendran P, Croteau-Chonka DC, Robins C, Torralbo A, Tomlinson C, Grünschläger F, Fitzpatrick N, Ytsma C, Kanno T, Gade S, Freitag D, Ziebell F, Haas S, Denaxas S, Betts JC, Wareham NJ, Hemingway H, Scott RA, Langenberg C. Proteomic signatures improve risk prediction for common and rare diseases. Nat Med 30: 2489–2498, 2024. doi:10.1038/s41591-024-03142-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 520. Argentieri MA, Xiao S, Bennett D, Winchester L, Nevado-Holgado AJ, Ghose U, Albukhari A, Yao P, Mazidi M, Lv J, Millwood I, Fry H, Rodosthenous RS, Partanen J, Zheng Z, Kurki M, Daly MJ, Palotie A, Adams CJ, Li L, Clarke R, Amin N, Chen Z, van Duijn CM. Proteomic aging clock predicts mortality and risk of common age-related diseases in diverse populations. Nat Med 30: 2450–2460, 2024. doi:10.1038/s41591-024-03164-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 521. Carrasco-Zanini J, Pietzner M, Koprulu M, Wheeler E, Kerrison ND, Wareham NJ, Langenberg C. Proteomic prediction of diverse incident diseases: a machine learning-guided biomarker discovery study using data from a prospective cohort study. Lancet Digit Health 6: e470–e479, 2024. doi:10.1016/S2589-7500(24)00087-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 522. Lehallier B, Gate D, Schaum N, Nanasi T, Lee SE, Yousef H, Moran Losada P, Berdnik D, Keller A, Verghese J, Sathyan S, Franceschi C, Milman S, Barzilai N, Wyss-Coray T. Undulating changes in human plasma proteome profiles across the lifespan. Nat Med 25: 1843–1850, 2019. doi:10.1038/s41591-019-0673-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 523. Shen X, Wang C, Zhou X, Zhou W, Hornburg D, Wu S, Snyder MP. Nonlinear dynamics of multi-omics profiles during human aging. Nat Aging 4: 1619–1634, 2024. doi:10.1038/s43587-024-00692-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 524. Williams SA, Kivimaki M, Langenberg C, Hingorani AD, Casas JP, Bouchard C, Jonasson C, Sarzynski MA, Shipley MJ, Alexander L, Ash J, Bauer T, Chadwick J, Datta G, DeLisle RK, Hagar Y, Hinterberg M, Ostroff R, Weiss S, Ganz P, Wareham NJ. Plasma protein patterns as comprehensive indicators of health. Nat Med 25: 1851–1857, 2019. doi:10.1038/s41591-019-0665-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 525. Guo Y, You J, Zhang Y, Liu WS, Huang YY, Zhang YR, Zhang W, Dong Q, Feng JF, Cheng W, Yu JT. Plasma proteomic profiles predict future dementia in healthy adults. Nat Aging 4: 247–260, 2024. doi:10.1038/s43587-023-00565-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 526. Carrasco-Zanini J, Wheeler E, Uluvar B, Kerrison N, Koprulu M, Wareham NJ, Pietzner M, Langenberg C. Mapping biological influences on the human plasma proteome beyond the genome. Nat Metab 6: 2010–2023, 2024. doi:10.1038/s42255-024-01133-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 527. Lind L, Mazidi M, Clarke R, Bennett DA, Zheng R. Measured and genetically predicted protein levels and cardiovascular diseases in UK Biobank and China Kadoorie Biobank. Nat Cardiovasc Res 3: 1189–1198, 2024. doi:10.1038/s44161-024-00545-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 528. Oh HS, Le Guen Y, Rappoport N, Urey DY, Rutledge J, Brunet A, Greicius MD, Wyss-Coray T. Plasma proteomics in the UK Biobank reveals youthful brains and immune systems promote healthspan and longevity. bioRxiv, 2024. doi:10.1101/2024.06.07.597771.
    Google Scholar
  • 529. Kivimäki M, Frank P, Pentti J, Jokela M, Nyberg ST, Blake A, Lindbohm JV, Oh HS-H, Singh-Manoux A, Wyss-Coray T, Partridge L. Proteomic organ-specific ageing signatures and 20-year risk of age-related diseases: the Whitehall II observational cohort study. Lancet Digit Health 7: e195–e204, 2025. doi:10.1016/j.landig.2025.01.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 530. Topol EJ. The revolution in high-throughput proteomics and AI. Science 385: eads5749, 2024. doi:10.1126/science.ads5749.
    Crossref | PubMed | Google Scholar
  • 531. Franks PW, Coral DE. Causal drivers of human proteome variation in health and disease. Nat Metab 6: 1854–1855, 2024. doi:10.1038/s42255-024-01138-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 532. Huang H, Chen Y, Xu W, Cao L, Qian K, Bischof E, Kennedy BK, Pu J. Decoding aging clocks: New insights from metabolomics. Cell Metab 37: 34–58, 2025. doi:10.1016/j.cmet.2024.11.007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 533. Yao S, Colangelo LA, Perry AS, Marron MM, Yaffe K, Sedaghat S, Lima JA, Tian Q, Clish CB, Newman AB, Shah RV, Murthy VL. Implications of metabolism on multi-systems healthy aging across the lifespan. Aging Cell 23: e14090, 2024. doi:10.1111/acel.14090.
    Crossref | PubMed | Web of Science | Google Scholar
  • 534. Vaiserman A, Krasnienkov D. Telomere length as a marker of biological age: state-of-the-art, open issues, and future perspectives. Front Genet 11: 630186, 2020. doi:10.3389/fgene.2020.630186.
    Crossref | PubMed | Web of Science | Google Scholar
  • 535. Daios S, Anogeianaki A, Kaiafa G, Kontana A, Veneti S, Gogou C, Karlafti E, Pilalas D, Kanellos I, Savopoulos C. Telomere length as a marker of biological aging: a critical review of recent literature. Curr Med Chem 29: 5478–5495, 2022. doi:10.2174/0929867329666220713123750.
    Crossref | PubMed | Web of Science | Google Scholar
  • 536. Sehgal R, Borrus D, Kasamato J, Armstrong JF, Gonzalez J, Markov Y, Priyanka A, Smith R, Carreras N, Dwaraka VB, Higgins-Chen A; DNAm aging biomarkers community, community Longevity interventional studies. DNAm aging biomarkers are responsive: insights from 51 longevity interventional studies in humans (Preprint). bioRxiv 2024.10.22.619522, 2024. doi:10.1101/2024.10.22.619522.
    Google Scholar
  • 537. Ying K, Paulson S, Reinhard J, de Lima Camillo LP, Trauble J, Jokiel S, Gobel D, Herzog C, Poganik JR, Moqri M, Gladyshev VN; Biomarkers of Aging Consortium. An open competition for biomarkers of aging (Preprint). bioRxiv 2024.10.29.620782, 2024. doi:10.1101/2024.10.29.620782.
    Google Scholar
  • 538. Waziry R, Ryan CP, Corcoran DL, Huffman KM, Kobor MS, Kothari M, Graf GH, Kraus VB, Kraus WE, Lin DT, Pieper CF, Ramaker ME, Bhapkar M, Das SK, Ferrucci L, Hastings WJ, Kebbe M, Parker DC, Racette SB, Shalev I, Schilling B, Belsky DW. Effect of long-term caloric restriction on DNA methylation measures of biological aging in healthy adults from the CALERIE trial. Nat Aging 3: 248–257, 2023. doi:10.1038/s43587-022-00357-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 539. McEwen LM, Jones MJ, Lin DT, Edgar RD, Husquin LT, MacIsaac JL, Ramadori KE, Morin AM, Rider CF, Carlsten C, Quintana-Murci L, Horvath S, Kobor MS. Systematic evaluation of DNA methylation age estimation with common preprocessing methods and the Infinium MethylationEPIC BeadChip array. Clin Epigenetics 10: 123, 2018. doi:10.1186/s13148-018-0556-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 540. Higgins-Chen AT, Thrush KL, Wang Y, Minteer CJ, Kuo PL, Wang M, Niimi P, Sturm G, Lin J, Moore AZ, Bandinelli S, Vinkers CH, Vermetten E, Rutten BP, Geuze E, Okhuijsen-Pfeifer C, van der Horst MZ, Schreiter S, Gutwinski S, Luykx JJ, Picard M, Ferrucci L, Crimmins EM, Boks MP, Hägg S, Hu-Seliger TT, Levine ME. A computational solution for bolstering reliability of epigenetic clocks: Implications for clinical trials and longitudinal tracking. Nat Aging 2: 644–661, 2022. doi:10.1038/s43587-022-00248-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 541. Nie C, Li Y, Li R, Yan Y, Zhang D, Li T, Li Z, Sun Y, Zhen H, Ding J, Wan Z, Gong J, Shi Y, Huang Z, Wu Y, Cai K, Zong Y, Wang Z, Wang R, Jian M, Jin X, Wang J, Yang H, Han JJ, Zhang X, Franceschi C, Kennedy BK, Xu X. Distinct biological ages of organs and systems identified from a multi-omics study. Cell Rep 38: 110459, 2022. doi:10.1016/j.celrep.2022.110459.
    Crossref | PubMed | Web of Science | Google Scholar
  • 542. Marttila S, Rajić S, Ciantar J, Mak JK, Junttila IS, Kummola L, Hägg S, Raitoharju E, Kananen L. Biological aging of different blood cell types. Geroscience 47: 1075–1092, 2025. doi:10.1007/s11357-024-01287-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 543. Nikopoulou C, Kleinenkuhnen N, Parekh S, Sandoval T, Ziegenhain C, Schneider F, Giavalisco P, Donahue KF, Vesting AJ, Kirchner M, Bozukova M, Vossen C, Altmüller J, Wunderlich T, Sandberg R, Kondylis V, Tresch A, Tessarz P. Spatial and single-cell profiling of the metabolome, transcriptome and epigenome of the aging mouse liver. Nat Aging 3: 1430–1445, 2023. doi:10.1038/s43587-023-00513-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 544. Sillanpää E, Heikkinen A, Kankaanpää A, Paavilainen A, Kujala UM, Tammelin TH, Kovanen V, Sipilä S, Pietiläinen KH, Kaprio J, Ollikainen M, Laakkonen EK. Blood and skeletal muscle ageing determined by epigenetic clocks and their associations with physical activity and functioning. Clin Epigenetics 13: 110, 2021. doi:10.1186/s13148-021-01094-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 545. Koncevičius K, Nair A, Šveikauskaitė A, Šeštokaitė A, Kazlauskaitė A, Dulskas A, Petronis A. Epigenetic age oscillates during the day. Aging Cell 23: e14170, 2024. doi:10.1111/acel.14170.
    Crossref | PubMed | Web of Science | Google Scholar
  • 546. Kerepesi C, Meer MV, Ablaeva J, Amoroso VG, Lee SG, Zhang B, Gerashchenko MV, Trapp A, Yim SH, Lu AT, Levine ME, Seluanov A, Horvath S, Park TJ, Gorbunova V, Gladyshev VN. Epigenetic aging of the demographically non-aging naked mole-rat. Nat Commun 13: 355, 2022. doi:10.1038/s41467-022-27959-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 547. Poganik JR, Zhang B, Baht GS, Tyshkovskiy A, Deik A, Kerepesi C, Yim SH, Lu AT, Haghani A, Gong T, Hedman AM, Andolf E, Pershagen G, Almqvist C, Clish CB, Horvath S, White JP, Gladyshev VN. Biological age is increased by stress and restored upon recovery. Cell Metab 35: 807–820.e5, 2023. doi:10.1016/j.cmet.2023.03.015.
    Crossref | PubMed | Web of Science | Google Scholar
  • 548. Moqri M, Poganik JR, Horvath S, Gladyshev VN. What makes biological age epigenetic clocks tick. Nat Aging 5: 335–336, 2025. doi:10.1038/s43587-025-00833-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 549. May M. Biomarkers of aging remain elusive as researchers try to slow the biological clock. Nat Med 29: 2673–2676, 2023. doi:10.1038/s41591-023-02560-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 550. Ledford H. How quickly are you ageing? What molecular ‘clocks’ can tell you about your health. Nature 638: 874–876, 2025. doi:10.1038/d41586-025-00566-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 551. Moqri M, Herzog C, Poganik JR, Ying K, Justice JN, Belsky DW, Higgins-Chen AT, Chen BH, Cohen AA, Fuellen G, Hägg S, Marioni RE, Widschwendter M, Fortney K, Fedichev PO, Zhavoronkov A, Barzilai N, Lasky-Su J, Kiel DP, Kennedy BK, Cummings S, Slagboom PE, Verdin E, Maier AB, Sebastiano V, Snyder MP, Gladyshev VN, Horvath S, Ferrucci L. Validation of biomarkers of aging. Nat Med 30: 360–372, 2024. doi:10.1038/s41591-023-02784-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 552. Prattichizzo F, Frigé C, Pellegrini V, Scisciola L, Santoro A, Monti D, Rippo MR, Ivanchenko M, Olivieri F, Franceschi C. Organ-specific biological clocks: ageotyping for personalized anti-aging medicine. Ageing Res Rev 96: 102253, 2024. doi:10.1016/j.arr.2024.102253.
    Crossref | PubMed | Web of Science | Google Scholar
  • 553. Jacques M, Landen S, Sharples AP, Garnham A, Schittenhelm R, Stele J, , et al.. Molecular landscape of modality-specific exercise adaptation in human skeletal muscle through large-scale multi-OMICs integration (Preprint). bioRxiv 2024.07.14.603458, 2024. doi:10.1101/2024.07.14.603458.
    Google Scholar
  • 554. Deng YT, You J, He Y, Zhang Y, Li HY, Wu XR, Cheng JY, Guo Y, Long ZW, Chen YL, Li ZY, Yang L, Zhang YR, Chen SD, Ge YJ, Huang YY, Shi LM, Dong Q, Mao Y, Feng JF, Cheng W, Yu JT. Atlas of the plasma proteome in health and disease in 53,026 adults. Cell 188: 253–271.e7, 2025. doi:10.1016/j.cell.2024.10.045.
    Crossref | PubMed | Web of Science | Google Scholar
  • 555. Nightingale Health Biobank Collaborative Group. Metabolomic and genomic prediction of common diseases in 700,217 participants in three national biobanks. Nat Commun 15: 10092, 2024. doi:10.1038/s41467-024-54357-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 556. Ren J, Song M, Zhang W, Cai JP, Cao F, Cao Z, , et al.. The Aging Biomarker Consortium represents a new era for aging research in China. Nat Med 29: 2162–2165, 2023. doi:10.1038/s41591-023-02444-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 557. Lyu YX, Fu Q, Wilczok D, Ying K, King A, Antebi A, , et al.. Longevity biotechnology: bridging AI, biomarkers, geroscience and clinical applications for healthy longevity. Aging (Albany NY) 16: 12955–12976, 2024. doi:10.18632/aging.206135.
    Crossref | PubMed | Google Scholar
  • 558. Marino N, Putignano G, Cappilli S, Chersoni E, Santuccione A, Calabrese G, Bischof E, Vanhaelen Q, Zhavoronkov A, Scarano B, Mazzotta AD, Santus E. Towards AI-driven longevity research: an overview. Front Aging 4: 1057204, 2023. doi:10.3389/fragi.2023.1057204.
    Crossref | PubMed | Web of Science | Google Scholar
  • 559. McGreevy KM, Radak Z, Torma F, Jokai M, Lu AT, Belsky DW, Binder A, Marioni RE, Ferrucci L, Pośpiech E, Branicki W, Ossowski A, Sitek A, Spólnicka M, Raffield LM, Reiner AP, Cox S, Kobor M, Corcoran DL, Horvath S. DNAmFitAge: biological age indicator incorporating physical fitness. Aging (Albany NY) 15: 3904–3938, 2023. doi:10.18632/aging.204538.
    Crossref | PubMed | Web of Science | Google Scholar
  • 560. Martínez-Magaña JJ, Hurtado-Soriano J, Rivero-Segura NA, Montalvo-Ortiz JL, Garcia-delaTorre P, Becerril-Rojas K, Gomez-Verjan JC. Towards a novel frontier in the use of epigenetic clocks in epidemiology. Arch Med Res 55: 103033, 2024. doi:10.1016/j.arcmed.2024.103033.
    Crossref | PubMed | Web of Science | Google Scholar
  • 561. Harris KM, Levitt B, Gaydosh L, Martin C, Meyer JM, Mishra AA, Kelly AL, Aiello AE. Sociodemographic and lifestyle factors and epigenetic aging in US young adults: NIMHD Social Epigenomics Program. JAMA Netw Open 7: e2427889, 2024. doi:10.1001/jamanetworkopen.2024.27889.
    Crossref | PubMed | Web of Science | Google Scholar
  • 562. Fong S, Pabis K, Latumalea D, Dugersuren N, Unfried M, Tolwinski N, Kennedy B, Gruber J. Principal component-based clinical aging clocks identify signatures of healthy aging and targets for clinical intervention. Nat Aging 4: 1137–1152, 2024. doi:10.1038/s43587-024-00646-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 563. Reicher L, Bar N, Godneva A, Reisner Y, Zahavi L, Shahaf N, Dhir R, Weinberger A, Segal E. Phenome-wide associations of human aging uncover sex-specific dynamics. Nat Aging 4: 1643–1655, 2024. doi:10.1038/s43587-024-00734-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 564. Horvath S, Topol EJ. Digitising the ageing process with epigenetic clocks. Lancet 404: 423, 2024. doi:10.1016/S0140-6736(24)01554-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 565. Grolaux R, Jones-Freeman B, Jacques M, Eynon N. The benefits of exercise on aging: focus on muscle biomarkers. Aging (Albany NY) 16: 11482–11483, 2024. doi:10.18632/aging.206064.
    Crossref | PubMed | Google Scholar
  • 566. Polidori MC. Aging hallmarks, biomarkers, and clocks for personalized medicine: (re)positioning the limelight. Free Radic Biol Med 215: 48–55, 2024. doi:10.1016/j.freeradbiomed.2024.02.012.
    Crossref | PubMed | Web of Science | Google Scholar
  • 567. Föhr T, Törmäkangas T, Lankila H, Viljanen A, Rantanen T, Ollikainen M, Kaprio J, Sillanpää E. The association between epigenetic clocks and physical functioning in older women: a 3-year follow-up. J Gerontol A Biol Sci Med Sci 77: 1569–1576, 2022. doi:10.1093/gerona/glab270.
    Crossref | PubMed | Web of Science | Google Scholar
  • 568. Frangos E, Graf C, Samaras N. Functional aging: integrating functionality to a multidimensional assessment of healthy aging. Curr Gerontol Geriatr Res 2023: 9409918, 2023. doi:10.1155/2023/9409918.
    Crossref | PubMed | Google Scholar
  • 569. Petnehazy N, Barnes HN, Newman AB, Kritchevsky SB, Cummings SR, Hepplen RT, Cawthon PM. Muscle mass, strength, power and physical performance and their association with quality of life in older adults, the Study of Muscle, Mobility and Aging (SOMMA). J Frailty Aging 13: 384–390, 2024. doi:10.14283/jfa.2024.45.
    Crossref | PubMed | Web of Science | Google Scholar
  • 570. Guarente L, Sinclair DA, Kroemer G. Human trials exploring anti-aging medicines. Cell Metab 36: 354–376, 2024. doi:10.1016/j.cmet.2023.12.007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 571. Mandsager K, Harb S, Cremer P, Phelan D, Nissen SE, Jaber W. Association of cardiorespiratory fitness with long-term mortality among adults undergoing exercise treadmill testing. JAMA Netw Open 1: e183605, 2018. doi:10.1001/jamanetworkopen.2018.3605.
    Crossref | PubMed | Web of Science | Google Scholar
  • 572. Kodama S, Saito K, Tanaka S, Maki M, Yachi Y, Asumi M, Sugawara A, Totsuka K, Shimano H, Ohashi Y, Yamada N, Sone H. Cardiorespiratory fitness as a quantitative predictor of all-cause mortality and cardiovascular events in healthy men and women: a meta-analysis. JAMA 301: 2024–2035, 2009. doi:10.1001/jama.2009.681.
    Crossref | PubMed | Web of Science | Google Scholar
  • 573. Kokkinos P, Faselis C, Samuel IB, Pittaras A, Doumas M, Murphy R, Heimall MS, Sui X, Zhang J, Myers J. Cardiorespiratory fitness and mortality risk across the spectra of age, race, and sex. J Am Coll Cardiol 80: 598–609, 2022. doi:10.1016/j.jacc.2022.05.031.
    Crossref | PubMed | Web of Science | Google Scholar
  • 574. Strasser B, Burtscher M. Survival of the fittest: VO2max, a key predictor of longevity? Front Biosci (Landmark Ed) 23: 1505–1516, 2018. doi:10.2741/4657.
    Crossref | PubMed | Web of Science | Google Scholar
  • 575. Blair SN, Kohl HW 3rd, Paffenbarger RS Jr, Clark DG, Cooper KH, Gibbons LW. Physical fitness and all-cause mortality. a prospective study of healthy men and women. JAMA 262: 2395–2401, 1989. doi:10.1001/jama.262.17.2395.
    Crossref | PubMed | Web of Science | Google Scholar
  • 576. Lang JJ, Prince SA, Merucci K, Cadenas-Sanchez C, Chaput JP, Fraser BJ, Manyanga T, McGrath R, Ortega FB, Singh B, Tomkinson GR. Cardiorespiratory fitness is a strong and consistent predictor of morbidity and mortality among adults: an overview of meta-analyses representing over 20.9 million observations from 199 unique cohort studies. Br J Sports Med 58: 556–566, 2024. doi:10.1136/bjsports-2023-107849.
    Crossref | PubMed | Web of Science | Google Scholar
  • 577. Lavie CJ, Sanchis-Gomar F, Ozemek C. Fit is it for longevity across populations. J Am Coll Cardiol 80: 610–612, 2022. doi:10.1016/j.jacc.2022.05.030.
    Crossref | PubMed | Web of Science | Google Scholar
  • 578. Sanchis-Gomar F, Lavie CJ, Marín J, Perez-Quilis C, Eijsvogels TM, O’Keefe JH, Perez MV, Blair SN. Exercise effects on cardiovascular disease: from basic aspects to clinical evidence. Cardiovasc Res 118: 2253–2266, 2022. doi:10.1093/cvr/cvab272.
    Crossref | PubMed | Web of Science | Google Scholar
  • 579. Schmid D, Leitzmann MF. Cardiorespiratory fitness as predictor of cancer mortality: a systematic review and meta-analysis. Ann Oncol 26: 272–278, 2015. doi:10.1093/annonc/mdu250.
    Crossref | PubMed | Web of Science | Google Scholar
  • 580. Sui X, Sarzynski MA, Lee DC, Kokkinos PF. Impact of changes in cardiorespiratory fitness on hypertension, dyslipidemia and survival: an overview of the epidemiological evidence. Prog Cardiovasc Dis 60: 56–66, 2017. doi:10.1016/j.pcad.2017.02.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 581. Franklin BA, Jae SY. Physical activity, cardiorespiratory fitness and atherosclerotic cardiovascular disease: part 1. Pulse (Basel) 12: 113–125, 2024. doi:10.1159/000541165.
    Crossref | PubMed | Web of Science | Google Scholar
  • 582. Franklin BA, Jae SY. Physical activity, cardiorespiratory fitness, and atherosclerotic cardiovascular disease: part 2. Pulse (Basel) 12: 126–138, 2024. doi:10.1159/000541166.
    Crossref | PubMed | Web of Science | Google Scholar
  • 583. Al-Mallah MH, Sakr S, Al-Qunaibet A. Cardiorespiratory fitness and cardiovascular disease prevention: an update. Curr Atheroscler Rep 20: 1, 2018. doi:10.1007/s11883-018-0711-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 584. Hill AV, Lupton H. Muscular exercise, lactic acid, and the supply and utilization of oxygen. Q J Med os-16: 135–171, 1923. doi:10.1093/qjmed/os-16.62.135.
    Crossref | Google Scholar
  • 585. Millet GP, Burtscher J, Bourdillon N, Manferdelli G, Burtscher M, Sandbakk Ø. The VO2max legacy of Hill and Lupton (1923)—100 years on. Int J Sports Physiol Perform 18: 1362–1365, 2023. doi:10.1123/ijspp.2023-0229.
    Crossref | PubMed | Web of Science | Google Scholar
  • 586. Bonikowske AR, Taylor JL, Larson KF, Hardwick J, Ozemek C, Harber MP, Kaminsky LA, Arena R, Lavie CJ. Evaluating current assessment techniques of cardiorespiratory fitness. Expert Rev Cardiovasc Ther 22: 231–241, 2024. doi:10.1080/14779072.2024.2363393.
    Crossref | PubMed | Web of Science | Google Scholar
  • 587. Smirmaul BP, Bertucci DR, Teixeira IP. Is the VO2max that we measure really maximal? Front Physiol 4: 203, 2013. doi:10.3389/fphys.2013.00203.
    Crossref | PubMed | Web of Science | Google Scholar
  • 588. Poole DC, Jones AM. Measurement of the maximum oxygen uptake V̇o2max: V̇o2peak is no longer acceptable. J Appl Physiol (1985) 122: 997–1002, 2017. doi:10.1152/japplphysiol.01063.2016.
    Link | Web of Science | Google Scholar
  • 589. Glaab T, Taube C. Practical guide to cardiopulmonary exercise testing in adults. Respir Res 23: 9, 2022. doi:10.1186/s12931-021-01895-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 590. Nichols S, Taylor C, Ingle L. A clinician’s guide to cardiopulmonary exercise testing 2: test interpretation. Br J Hosp Med (Lond) 76: 281–289, 2015. doi:10.12968/hmed.2015.76.5.281.
    Crossref | PubMed | Web of Science | Google Scholar
  • 591. Taylor C, Nichols S, Ingle L. A clinician’s guide to cardiopulmonary exercise testing 1: an introduction. Br J Hosp Med (Lond) 76: 192–195, 2015. doi:10.12968/hmed.2015.76.4.192.
    Crossref | PubMed | Web of Science | Google Scholar
  • 592. Balady GJ, Arena R, Sietsema K, Myers J, Coke L, Fletcher GF, Forman D, Franklin B, Guazzi M, Gulati M, Keteyian SJ, Lavie CJ, Macko R, Mancini D, Milani RV; American Heart Association Exercise, Cardiac Rehabilitation, and Prevention Committee of the Council on Clinical Cardiology, Council on Epidemiology and Prevention, Council on Peripheral Vascular Disease, Interdisciplinary Council on Quality of Care and Outcomes Research. Clinician’s Guide to cardiopulmonary exercise testing in adults: a scientific statement from the American Heart Association. Circulation 122: 191–225, 2010. doi:10.1161/CIR.0b013e3181e52e69.
    Crossref | PubMed | Web of Science | Google Scholar
  • 593. Kaminsky LA, Arena R, Ellingsen O, Harber MP, Myers J, Ozemek C, Ross R. Cardiorespiratory fitness and cardiovascular—the past, present, and future. Prog Cardiovasc Dis 62: 86–93, 2019. doi:10.1016/j.pcad.2019.01.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 594. Williams N. The Borg Rating of Perceived Exertion (RPE) scale. Occup Med 67: 404–405, 2017. doi:10.1093/occmed/kqx063.
    Crossref | Web of Science | Google Scholar
  • 595. Ozemek C, Hardwick J, Bonikowske A, Christle J, German C, Reddy S, Arena R, Faghy M. How to interpret a cardiorespiratory fitness assessment—key measures that provide the best picture of health, disease status and prognosis. Prog Cardiovasc Dis 83: 23–28, 2024. doi:10.1016/j.pcad.2024.02.014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 596. McConnell TR. Practical considerations in the testing of VO2max in runners. Sports Med 5: 57–68, 1988. doi:10.2165/00007256-198805010-00005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 597. Price S, Wiecha S, Cieslinski I, Sliz D, Kasiak PS, Lach J, Gruba G, Kowalski T, Mamcarz A. Differences between treadmill and cycle ergometer cardiopulmonary exercise testing results in triathletes and their association with body composition and body mass index. Int J Environ Res Public Health 19: 3557, 2022. doi:10.3390/ijerph19063557.
    Crossref | PubMed | Web of Science | Google Scholar
  • 598. Lundby C, Montero D, Joyner M. Biology of VO2max: looking under the physiology lamp. Acta Physiol (Oxf) 220: 218–228, 2017. doi:10.1111/apha.12827.
    Crossref | PubMed | Web of Science | Google Scholar
  • 599. Mello ESF, Oliveira A, Santanna TD, Soares P, Rodrigues GD. Updates in inspiratory muscle training for older adults: a systematic review. Arch Gerontol Geriatr 127: 105579, 2024. doi:10.1016/j.archger.2024.105579.
    Crossref | PubMed | Web of Science | Google Scholar
  • 600. Hawkins S, Wiswell R. Rate and mechanism of maximal oxygen consumption decline with aging: implications for exercise training. Sports Med 33: 877–888, 2003. doi:10.2165/00007256-200333120-00002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 601. Letnes JM, Nes BM, Wisløff U. Age-related decline in peak oxygen uptake: cross-sectional vs. longitudinal findings. A review. Int J Cardiol Cardiovasc Risk Prev 16: 200171, 2023. doi:10.1016/j.ijcrp.2023.200171.
    Crossref | PubMed | Web of Science | Google Scholar
  • 602. Lundby C, Robach P. Performance enhancement: what are the physiological limits? Physiology (Bethesda) 30: 282–292, 2015. doi:10.1152/physiol.00052.2014.
    Link | Web of Science | Google Scholar
  • 603. Morey MC, Pieper CF, Cornoni-Huntley J. Is there a threshold between peak oxygen uptake and self-reported physical functioning in older adults? Med Sci Sports Exerc 30: 1223–1229, 1998. doi:10.1097/00005768-199808000-00007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 604. Moored KD, Qiao YS, Rosso AL, Toledo FG, Cawthon PM, Cummings SR, Goodpaster BH, Kritchevsky SB, Glynn NW. Dual roles of cardiorespiratory fitness and fatigability in the life-space mobility of older adults: the Study of Muscle, Mobility and Aging (SOMMA). J Gerontol A Biol Sci Med Sci 78: 1392–1401, 2023. doi:10.1093/gerona/glad037.
    Crossref | PubMed | Web of Science | Google Scholar
  • 605. Ross R, Myers J. Cardiorespiratory fitness and its place in medicine. Rev Cardiovasc Med 24: 14, 2023. doi:10.31083/j.rcm2401014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 606. Laukkanen JA, Isiozor NM, Kunutsor SK. Objectively assessed cardiorespiratory fitness and all-cause mortality risk: an updated meta-analysis of 37 cohort studies involving 2,258,029 participants. Mayo Clin Proc 97: 1054–1073, 2022. doi:10.1016/j.mayocp.2022.02.029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 607. Kohl HW, Gordon NF, Villegas JA, Blair SN. Cardiorespiratory fitness, glycemic status, and mortality risk in men. Diabetes Care 15: 184–192, 1992. doi:10.2337/diacare.15.2.184.
    Crossref | PubMed | Web of Science | Google Scholar
  • 608. Gaesser GA, Angadi SS. Obesity treatment: weight loss versus increasing fitness and physical activity for reducing health risks. iScience 24: 102995, 2021. doi:10.1016/j.isci.2021.102995.
    Crossref | PubMed | Web of Science | Google Scholar
  • 609. Ortega FB, Lavie CJ, Blair SN. Obesity and cardiovascular disease. Circ Res 118: 1752–1770, 2016. doi:10.1161/CIRCRESAHA.115.306883.
    Crossref | PubMed | Web of Science | Google Scholar
  • 610. Barry VW, Baruth M, Beets MW, Durstine JL, Liu J, Blair SN. Fitness vs. fatness on all-cause mortality: a meta-analysis. Prog Cardiovasc Dis 56: 382–390, 2014. doi:10.1016/j.pcad.2013.09.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 611. Lee CD, Blair SN, Jackson AS. Cardiorespiratory fitness, body composition, and all-cause and cardiovascular disease mortality in men. Am J Clin Nutr 69: 373–380, 1999. doi:10.1093/ajcn/69.3.373.
    Crossref | PubMed | Web of Science | Google Scholar
  • 612. Weeldreyer NR, De Guzman JC, Paterson C, Allen JD, Gaesser GA, Angadi SS. Cardiorespiratory fitness, body mass index and mortality: a systematic review and meta-analysis. Br J Sports Med 59: 339–346, 2025. doi:10.1136/bjsports-2024-108748.
    Crossref | PubMed | Web of Science | Google Scholar
  • 613. Wang S, Xu L, Yang W, Wang J, Dove A, Qi X, Xu W. Association of cardiorespiratory fitness with dementia risk across different levels of genetic predisposition: a large community-based longitudinal study. Br J Sports Med 59: 150–158, 2025. doi:10.1136/bjsports-2023-108048.
    Crossref | PubMed | Web of Science | Google Scholar
  • 614. Kokkinos P, Faselis C, Franklin B, Lavie CJ, Sidossis L, Moore H, Karasik P, Myers J. Cardiorespiratory fitness, body mass index and heart failure incidence. Eur J Heart Fail 21: 436–444, 2019. doi:10.1002/ejhf.1433.
    Crossref | PubMed | Web of Science | Google Scholar
  • 615. Hoppeler H. Deciphering V̇O2,max: limits of the genetic approach. J Exp Biol 221: jeb164327, 2018. doi:10.1242/jeb.164327.
    Crossref | PubMed | Web of Science | Google Scholar
  • 616. Meyler S, Bottoms L, Muniz-Pumares D. Biological and methodological factors affecting V̇O2max response variability to endurance training and the influence of exercise intensity prescription. Exp Physiol 106: 1410–1424, 2021. doi:10.1113/EP089565.
    Crossref | PubMed | Web of Science | Google Scholar
  • 617. Rønnestad BR, Hansen J, Stensløkken L, Joyner MJ, Lundby C. Case Studies in Physiology: Temporal changes in determinants of aerobic performance in individual going from alpine skier to world junior champion time trial cyclist. J Appl Physiol (1985) 127: 306–311, 2019. doi:10.1152/japplphysiol.00798.2018.
    Link | Web of Science | Google Scholar
  • 618. Imboden MT, Harber MP, Whaley MH, Finch WH, Bishop DL, Fleenor BS, Kaminsky LA. The association between the change in directly measured cardiorespiratory fitness across time and mortality risk. Prog Cardiovasc Dis 62: 157–162, 2019. doi:10.1016/j.pcad.2018.12.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 619. Robinson AT, Watso JC, Babcock MC, Joyner MJ, Farquhar WB. Record-breaking performance in a 70-year-old marathoner. N Engl J Med 380: 1485–1486, 2019. doi:10.1056/NEJMc1900771.
    Crossref | PubMed | Web of Science | Google Scholar
  • 620. Gries KJ, Raue U, Perkins RK, Lavin KM, Overstreet BS, D’Acquisto LJ, Graham B, Finch WH, Kaminsky LA, Trappe TA, Trappe S. Cardiovascular and skeletal muscle health with lifelong exercise. J Appl Physiol (1985) 125: 1636–1645, 2018. doi:10.1152/japplphysiol.00174.2018.
    Link | Web of Science | Google Scholar
  • 621. Burtscher J, Strasser B, Burtscher M, Millet GP. The impact of training on the loss of cardiorespiratory fitness in aging masters endurance athletes. Int J Environ Res Public Health 19: 11050, 2022. doi:10.3390/ijerph191711050.
    Crossref | PubMed | Web of Science | Google Scholar
  • 622. Kokkinos P, Myers J. Cardiorespiratory fitness and health outcomes across the spectra of age, gender, and race. Rev Cardiovasc Med 25: 15, 2024. doi:10.31083/j.rcm2501015.
    Crossref | PubMed | Web of Science | Google Scholar
  • 623. Allen WK, Seals DR, Hurley BF, Ehsani AA, Hagberg JM. Lactate threshold and distance-running performance in young and older endurance athletes. J Appl Physiol (1985) 58: 1281–1284, 1985. doi:10.1152/jappl.1985.58.4.1281.
    Link | Web of Science | Google Scholar
  • 624. Valenzuela PL, Maffiuletti NA, Joyner MJ, Lucia A, Lepers R. Lifelong endurance exercise as a countermeasure against age-related V̇O2max decline: physiological overview and insights from masters athletes. Sports Med 50: 703–716, 2020. doi:10.1007/s40279-019-01252-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 625. Van Hooren B, Plasqui G, Lepers R. Physiological, spatiotemporal, anthropometric, training, and performance characteristics of a 75-year-old multiple world record holder middle-distance runner. Int J Sports Physiol Perform 18: 204–208, 2023. doi:10.1123/ijspp.2022-0284.
    Crossref | PubMed | Web of Science | Google Scholar
  • 626. Billat V, Dhonneur G, Mille-Hamard L, Le Moyec L, Momken I, Launay T, Koralsztein JP, Besse S. Case Studies in Physiology: Maximal oxygen consumption and performance in a centenarian cyclist. J Appl Physiol (1985) 122: 430–434, 2017. doi:10.1152/japplphysiol.00569.2016.
    Link | Web of Science | Google Scholar
  • 627. Chambers DJ, Wisely NA. Cardiopulmonary exercise testing—a beginner’s guide to the nine-panel plot. BJA Educ 19: 158–164, 2019. doi:10.1016/j.bjae.2019.01.009.
    Crossref | PubMed | Google Scholar
  • 628. Del Punta L, De Biase N, Armenia S, Di Fiore V, Maremmani D, Gargani L, Mazzola M, De Carlo M, Mengozzi A, Lomonaco T, Galeotti GG, Dini FL, Masi S, Pugliese NR. Combining cardiopulmonary exercise testing with echocardiography: a multiparametric approach to the cardiovascular and cardiopulmonary systems. Eur Heart J Imaging Methods Pract 1: qyad021, 2023. doi:10.1093/ehjimp/qyad021.
    Crossref | PubMed | Google Scholar
  • 629. Liu CM, Kuo MJ, Kuo CY, Wu IC, Chen PF, Hsu WT, Liao LL, Chen SA, Tsao HM, Liu CL, Hu YF. Reclassification of the conventional risk assessment for aging-related diseases by electrocardiogram-enabled biological age. NPJ Aging 11: 7, 2025. doi:10.1038/s41514-025-00198-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 630. Ghidoni C, Kruzik M, Rossi VA, Caselli S, Schmied CM, Niederseer D. Definitions for hypertensive response to exercise. Cardiol Rev 32: 273–278, 2024. doi:10.1097/CRD.0000000000000506.
    Crossref | PubMed | Web of Science | Google Scholar
  • 631. Schultz MG, Otahal P, Cleland VJ, Blizzard L, Marwick TH, Sharman JE. Exercise-induced hypertension, cardiovascular events, and mortality in patients undergoing exercise stress testing: a systematic review and meta-analysis. Am J Hypertens 26: 357–366, 2013. doi:10.1093/ajh/hps053.
    Crossref | PubMed | Web of Science | Google Scholar
  • 632. Radtke T, Crook S, Kaltsakas G, Louvaris Z, Berton D, Urquhart DS, Kampouras A, Rabinovich RA, Verges S, Kontopidis D, Boyd J, Tonia T, Langer D, De Brandt J, Goërtz YM, Burtin C, Spruit MA, Braeken DC, Dacha S, Franssen FM, Laveneziana P, Eber E, Troosters T, Neder JA, Puhan MA, Casaburi R, Vogiatzis I, Hebestreit H. ERS statement on standardisation of cardiopulmonary exercise testing in chronic lung diseases. Eur Respir Rev 28: 180101, 2019. doi:10.1183/16000617.0101-2018.
    Crossref | PubMed | Web of Science | Google Scholar
  • 633. Trankle CR, Canada JM, Jordan JH, Truong U, Hundley WG. Exercise cardiovascular magnetic resonance: a review. J Magn Reson Imaging 55: 720–754, 2022. doi:10.1002/jmri.27580.
    Crossref | PubMed | Web of Science | Google Scholar
  • 634. Jou J, Zhou X, Lindow T, Brudin L, Hedman K, Ekström M, Malinovschi A. Heart rate response and recovery in cycle exercise testing: normal values and association with mortality. Eur J Prev Cardiol 32: 32–42, 2025. doi:10.1093/eurjpc/zwae308.
    Crossref | PubMed | Web of Science | Google Scholar
  • 635. Cole CR, Blackstone EH, Pashkow FJ, Snader CE, Lauer MS. Heart-rate recovery immediately after exercise as a predictor of mortality. N Engl J Med 341: 1351–1357, 1999. doi:10.1056/NEJM199910283411804.
    Crossref | PubMed | Web of Science | Google Scholar
  • 636. Qiu S, Cai X, Sun Z, Li L, Zuegel M, Steinacker JM, Schumann U. Heart rate recovery and risk of cardiovascular events and all-cause mortality: a meta-analysis of prospective cohort studies. J Am Heart Assoc 6: e005505, 2017. doi:10.1161/JAHA.117.005505.
    Crossref | PubMed | Web of Science | Google Scholar
  • 637. Tanner V, Millet GP, Bourdillon N. Agreement between heart rate variability - derived vs. ventilatory and lactate thresholds: a systematic review with meta-analyses. Sports Med Open 10: 109, 2024. doi:10.1186/s40798-024-00768-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 638. Jarczok MN, Weimer K, Braun C, Williams DP, Thayer JF, Gündel HO, Balint EM. Heart rate variability in the prediction of mortality: a systematic review and meta-analysis of healthy and patient populations. Neurosci Biobehav Rev 143: 104907, 2022. doi:10.1016/j.neubiorev.2022.104907.
    Crossref | PubMed | Web of Science | Google Scholar
  • 639. Jain CC, Borlaug BA. Performance and interpretation of invasive hemodynamic exercise testing. Chest 158: 2119–2129, 2020. doi:10.1016/j.chest.2020.05.552.
    Crossref | PubMed | Web of Science | Google Scholar
  • 640. Faude O, Kindermann W, Meyer T. Lactate threshold concepts: how valid are they? Sports Med 39: 469–490, 2009. doi:10.2165/00007256-200939060-00003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 641. Poole DC, Rossiter HB, Brooks GA, Gladden LB. The anaerobic threshold: 50+ years of controversy. J Physiol 599: 737–767, 2021. doi:10.1113/JP279963.
    Crossref | PubMed | Web of Science | Google Scholar
  • 642. Joyner MJ, Hunter SK, Lucia A, Jones AM. Physiology and fast marathons. J Appl Physiol (1985) 128: 1065–1068, 2020. doi:10.1152/japplphysiol.00793.2019.
    Link | Web of Science | Google Scholar
  • 643. Jones AM. The fourth dimension: physiological resilience as an independent determinant of endurance exercise performance. J Physiol 602: 4113–4128, 2024. doi:10.1113/JP284205.
    Crossref | PubMed | Web of Science | Google Scholar
  • 644. Joyner MJ, Coyle EF. Endurance exercise performance: the physiology of champions. J Physiol 586: 35–44, 2008. doi:10.1113/jphysiol.2007.143834.
    Crossref | PubMed | Web of Science | Google Scholar
  • 645. Millet GP, Chamari K. Look to the stars—is there anything that public health and rehabilitation can learn from elite sports? Front Sports Act Living 4: 1072154, 2022. doi:10.3389/fspor.2022.1072154.
    Crossref | PubMed | Web of Science | Google Scholar
  • 646. McKay AK, Stellingwerff T, Smith ES, Martin DT, Mujika I, Goosey-Tolfrey VL, Sheppard J, Burke LM. Defining training and performance caliber: a participant classification framework. Int J Sports Physiol Perform 17: 317–331, 2022. doi:10.1123/ijspp.2021-0451.
    Crossref | PubMed | Web of Science | Google Scholar
  • 647. Burke LM, Whitfield J, Hawley JA. The race within a race: together on the marathon starting line but miles apart in the experience. Free Radic Biol Med 227: 367–378, 2025. doi:10.1016/j.freeradbiomed.2024.10.277.
    Crossref | PubMed | Web of Science | Google Scholar
  • 648. Harber MP, Kaminsky LA, Arena R, Blair SN, Franklin BA, Myers J, Ross R. Impact of cardiorespiratory fitness on all-cause and disease-specific mortality: advances since 2009. Prog Cardiovasc Dis 60: 11–20, 2017. doi:10.1016/j.pcad.2017.03.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 649. Kampert JB, Blair SN, Barlow CE, Kohl HW 3rd. Physical activity, physical fitness, and all-cause and cancer mortality: a prospective study of men and women. Ann Epidemiol 6: 452–457, 1996. doi:10.1016/s1047-2797(96)00059-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 650. Sui X, LaMonte MJ, Laditka JN, Hardin JW, Chase N, Hooker SP, Blair SN. Cardiorespiratory fitness and adiposity as mortality predictors in older adults. JAMA 298: 2507–2516, 2007. doi:10.1001/jama.298.21.2507.
    Crossref | PubMed | Web of Science | Google Scholar
  • 651. Sui X, Laditka JN, Hardin JW, Blair SN. Estimated functional capacity predicts mortality in older adults. J Am Geriatr Soc 55: 1940–1947, 2007. doi:10.1111/j.1532-5415.2007.01455.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 652. Myers J, Prakash M, Froelicher V, Do D, Partington S, Atwood JE. Exercise capacity and mortality among men referred for exercise testing. N Engl J Med 346: 793–801, 2002. doi:10.1056/NEJMoa011858.
    Crossref | PubMed | Web of Science | Google Scholar
  • 653. Blair SN, Kohl HW 3rd, Barlow CE, Paffenbarger RS Jr, Gibbons LW, Macera CA. Changes in physical fitness and all-cause mortality. A prospective study of healthy and unhealthy men. JAMA 273: 1093–1098, 1995. doi:10.1001/jama.1995.03520380029031.
    Crossref | PubMed | Web of Science | Google Scholar
  • 654. Gulati M, Pandey DK, Arnsdorf MF, Lauderdale DS, Thisted RA, Wicklund RH, Al-Hani AJ, Black HR. Exercise capacity and the risk of death in women: the St James Women Take Heart Project. Circulation 108: 1554–1559, 2003. doi:10.1161/01.CIR.0000091080.57509.E9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 655. Kokkinos P, Myers J, Faselis C, Panagiotakos DB, Doumas M, Pittaras A, Manolis A, Kokkinos JP, Karasik P, Greenberg M, Papademetriou V, Fletcher R. Exercise capacity and mortality in older men: a 20-year follow-up study. Circulation 122: 790–797, 2010. doi:10.1161/CIRCULATIONAHA.110.938852.
    Crossref | PubMed | Web of Science | Google Scholar
  • 656. Farrell SW, Leonard D, Barlow CE, Shuval K, Pavlovic A, Defina LF. Examining the gradient of all-cause mortality risk in women across the cardiorespiratory fitness continuum. Med Sci Sports Exerc 54: 1904–1910, 2022. doi:10.1249/MSS.0000000000002988.
    Crossref | PubMed | Web of Science | Google Scholar
  • 657. Erikssen G, Liestøl K, Bjornholt J, Thaulow E, Sandvik L, Erikssen J. Changes in physical fitness and changes in mortality. Lancet 352: 759–762, 1998. doi:10.1016/S0140-6736(98)02268-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 658. Jackson AS, Sui X, O’Connor DP, Church TS, Lee DC, Artero EG, Blair SN. Longitudinal cardiorespiratory fitness algorithms for clinical settings. Am J Prev Med 43: 512–519, 2012. doi:10.1016/j.amepre.2012.06.032.
    Crossref | PubMed | Web of Science | Google Scholar
  • 659. Artero EG, Jackson AS, Sui X, Lee DC, O’Connor DP, Lavie CJ, Church TS, Blair SN. Longitudinal algorithms to estimate cardiorespiratory fitness: associations with nonfatal cardiovascular disease and disease-specific mortality. J Am Coll Cardiol 63: 2289–2296, 2014. doi:10.1016/j.jacc.2014.03.008.
    Crossref | PubMed | Web of Science | Google Scholar
  • 660. Peterman JE, Whaley MH, Harber MP, Fleenor BS, Imboden MT, Myers J, Arena R, Kaminsky LA. Comparison of non-exercise cardiorespiratory fitness prediction equations in apparently healthy adults. Eur J Prev Cardiol 28: 142–148–148, 2021. doi:10.1177/2047487319881242.
    Crossref | PubMed | Web of Science | Google Scholar
  • 661. Cabanas-Sánchez V, Artero EG, Lavie CJ, Higueras-Fresnillo S, García-Esquinas E, Sadarangani KP, Ortolá R, Rodríguez-Artalejo F, Martínez-Gómez D. Prediction of cardiovascular health by non-exercise estimated cardiorespiratory fitness. Heart 106: 1832–1838, 2020. doi:10.1136/heartjnl-2020-316871.
    Crossref | PubMed | Web of Science | Google Scholar
  • 662. Lee I, Kim S, Kang H. Non-exercise based estimation of cardiorespiratory fitness is inversely associated with metabolic syndrome in a representative sample of Korean adults. BMC Geriatr 20: 146, 2020. doi:10.1186/s12877-020-01558-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 663. Zhu YJ, Fu WR, Lu WJ, Wang XL, Wang X, Shan YG, Zheng XL, Li R, Peng M, Pan L, Qiu J, Qin XF, Sun GJ, Wang L, Dong JZ, Xiao LL, Qiu CG. Non-exercise estimated cardiorespiratory fitness and mortality among adults with hypertension. Am J Hypertens 38: 63–71, 2024. doi:10.1093/ajh/hpae137.
    Crossref | PubMed | Web of Science | Google Scholar
  • 664. Wicks JR, Oldridge NB, Nielsen LK, Vickers CE. HR index—a simple method for the prediction of oxygen uptake. Med Sci Sports Exerc 43: 2005–2012, 2011. doi:10.1249/MSS.0b013e318217276e.
    Crossref | PubMed | Web of Science | Google Scholar
  • 665. Standl E, Erbach M, Schnell O. Defending the con side: obesity paradox does not exist. Diabetes Care 36, Suppl 2: S282–S286, 2013. doi:10.2337/dcS13-2040.
    Crossref | PubMed | Web of Science | Google Scholar
  • 666. Hainer V, Aldhoon-Hainerová I. Obesity paradox does exist. Diabetes Care 36, Suppl 2: S276–S281, 2013. doi:10.2337/dcS13-2023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 667. Sato R, von Haehling S. Revisiting the obesity paradox in heart failure: what is the best anthropometric index to gauge obesity? Eur Heart J 44: 1154–1156, 2023. doi:10.1093/eurheartj/ehad079.
    Crossref | PubMed | Web of Science | Google Scholar
  • 668. Prado CM, Gonzalez MC, Heymsfield SB. Body composition phenotypes and obesity paradox. Curr Opin Clin Nutr Metab Care 18: 535–551, 2015. doi:10.1097/MCO.0000000000000216.
    Crossref | PubMed | Web of Science | Google Scholar
  • 669. Aune D, Sen A, Prasad M, Norat T, Janszky I, Tonstad S, Romundstad P, Vatten LJ. BMI and all cause mortality: systematic review and non-linear dose-response meta-analysis of 230 cohort studies with 3.74 million deaths among 30.3 million participants. BMJ 353: i2156, 2016. doi:10.1136/bmj.i2156.
    Crossref | PubMed | Web of Science | Google Scholar
  • 670. Lucas E, Aronne LJ. Is it time to define obesity by body composition and not solely body mass index? J Clin Endocrinol Metab 110: e1278–e1279, 2025. doi:10.1210/clinem/dgae473.
    Crossref | PubMed | Web of Science | Google Scholar
  • 671. Lee DH, Keum N, Hu FB, Orav EJ, Rimm EB, Willett WC, Giovannucci EL. Predicted lean body mass, fat mass, and all cause and cause specific mortality in men: prospective US cohort study. BMJ 362: k2575, 2018. doi:10.1136/bmj.k2575.
    Crossref | PubMed | Web of Science | Google Scholar
  • 672. Rubino F, Cummings DE, Eckel RH, Cohen RV, Wilding JP, Brown WA, , et al.. Definition and diagnostic criteria of clinical obesity. Lancet Diabetes Endocrinol 13: 221–262, 2025. doi:10.1016/S2213-8587(24)00316-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 673. Alcazar J, Navarrete-Villanueva D, Mañas A, Gómez-Cabello A, Pedrero-Chamizo R, Alegre LM, Villa G, Gusi N, González-Gross M, Casajús JA, Vicente-Rodriguez G, Ara I. ‘Fat but powerful’ paradox: association of muscle power and adiposity markers with all-cause mortality in older adults from the EXERNET multicentre study. Br J Sports Med 55: 1204–1211, 2021. doi:10.1136/bjsports-2020-103720.
    Crossref | PubMed | Web of Science | Google Scholar
  • 674. Masters RK. Sources and severity of bias in estimates of the BMI-mortality association. Popul Stud (Camb) 77: 35–53, 2023. doi:10.1080/00324728.2023.2168035.
    Crossref | PubMed | Web of Science | Google Scholar
  • 675. Srikanthan P, Karlamangla AS. Muscle mass index as a predictor of longevity in older adults. Am J Med 127: 547–553, 2014. doi:10.1016/j.amjmed.2014.02.007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 676. Kim D, Lee J, Park R, Oh CM, Moon S. Association of low muscle mass and obesity with increased all-cause and cardiovascular disease mortality in US adults. J Cachexia Sarcopenia Muscle 15: 240–254, 2024. doi:10.1002/jcsm.13397.
    Crossref | PubMed | Web of Science | Google Scholar
  • 677. Wang DX, Yao J, Zirek Y, Reijnierse EM, Maier AB. Muscle mass, strength, and physical performance predicting activities of daily living: a meta-analysis. J Cachexia Sarcopenia Muscle 11: 3–25, 2020. doi:10.1002/jcsm.12502.
    Crossref | PubMed | Web of Science | Google Scholar
  • 678. Ceolin C, Acunto V, Simonato C, Cazzavillan S, Vergadoro M, Papa MV, Trapella GS, Sermasi R, Noale M, De Rui M, Zanforlini BM, Curreri C, Bertocco A, Devita M, Coin A, Sergi G. New perspectives in the association between anthropometry and mortality: the role of calf circumference. J Frailty Aging 13: 108–115, 2024. doi:10.14283/jfa.2024.4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 679. Prado CM, Purcell SA, Alish C, Pereira SL, Deutz NE, Heyland DK, Goodpaster BH, Tappenden KA, Heymsfield SB. Implications of low muscle mass across the continuum of care: a narrative review. Ann Med 50: 675–693, 2018. doi:10.1080/07853890.2018.1511918.
    Crossref | PubMed | Web of Science | Google Scholar
  • 680. Santanasto AJ, Goodpaster BH, Kritchevsky SB, Miljkovic I, Satterfield S, Schwartz AV, Cummings SR, Boudreau RM, Harris TB, Newman AB. Body composition remodeling and mortality: the Health Aging and Body Composition Study. J Gerontol A Biol Sci Med Sci 72: 513–519, 2017. doi:10.1093/gerona/glw163.
    Crossref | PubMed | Web of Science | Google Scholar
  • 681. González-Torres LA, Acuña-Rocha VD, Herrera-Nuñez M, Millán-Alanís JM, de la Cruz-de la Cruz C, Cruz-López RH, Alcalá-González JI, Moreno-Hoyos-Abril JF, Gonzalez-Aguirre JE. Low muscle mass in the internal medicine ward: prevalence and survival implications. Arch Med Res 56: 103103, 2025. doi:10.1016/j.arcmed.2024.103103.
    Crossref | PubMed | Web of Science | Google Scholar
  • 682. Linge J, Petersson M, Forsgren MF, Sanyal AJ, Dahlqvist Leinhard O. Adverse muscle composition predicts all-cause mortality in the UK Biobank imaging study. J Cachexia Sarcopenia Muscle 12: 1513–1526, 2021. doi:10.1002/jcsm.12834.
    Crossref | PubMed | Web of Science | Google Scholar
  • 683. Liu M, Zhang Z, Zhou C, Ye Z, He P, Zhang Y, Li H, Liu C, Qin X. Predicted fat mass and lean mass in relation to all-cause and cause-specific mortality. J Cachexia Sarcopenia Muscle 13: 1064–1075, 2022. doi:10.1002/jcsm.12921.
    Crossref | PubMed | Web of Science | Google Scholar
  • 684. Zhou HH, Liao Y, Peng Z, Liu F, Wang Q, Yang W. Association of muscle wasting with mortality risk among adults: a systematic review and meta-analysis of prospective studies. J Cachexia Sarcopenia Muscle 14: 1596–1612, 2023. doi:10.1002/jcsm.13263.
    Crossref | PubMed | Web of Science | Google Scholar
  • 685. Mariean CR, Tiucă OM, Mariean A, Cotoi OS. Cancer cachexia: new insights and future directions. Cancers (Basel) 15, 2023. doi:10.3390/cancers15235590.
    Crossref | PubMed | Web of Science | Google Scholar
  • 686. Ferrer M, Anthony TG, Ayres JS, Biffi G, Brown JC, Caan BJ, Cespedes Feliciano EM, Coll AP, Dunne RF, Goncalves MD, Grethlein J, Heymsfield SB, Hui S, Jamal-Hanjani M, Lam JM, Lewis DY, McCandlish D, Mustian KM, O'Rahilly S, Perrimon N, White EP, Janowitz T. Cachexia: a systemic consequence of progressive, unresolved disease. Cell 186: 1824–1845, 2023. doi:10.1016/j.cell.2023.03.028.
    Crossref | PubMed | Web of Science | Google Scholar
  • 687. Xu J, Wan CS, Ktoris K, Reijnierse EM, Maier AB. Sarcopenia is associated with mortality in adults: a systematic review and meta-analysis. Gerontology 68: 361–376, 2022. doi:10.1159/000517099.
    Crossref | PubMed | Web of Science | Google Scholar
  • 688. Koon-Yee Lee G, Chun-Ming Au P, Hoi-Yee Li G, Chan M, Li HL, Man-Yung Cheung B, Chi-Kei Wong I, Ho-Fun Lee V, Mok J, Hon-Kei Yip B, King-Yip Cheng K, Wu CH, Cheung CL. Sarcopenia and mortality in different clinical conditions: a meta-analysis. Osteoporos Sarcopenia 7: S19–S27, 2021. doi:10.1016/j.afos.2021.02.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 689. Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, Murphy R, Ghosh S, Sawyer MB, Baracos VE. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol 31: 1539–1547, 2013. doi:10.1200/JCO.2012.45.2722.
    Crossref | PubMed | Web of Science | Google Scholar
  • 690. Liu C, Zhang Q, Liu T, Zhang Q, Song M, Ruan G, Lin S, Wang Z, Zheng X, Chen Y, Zhang H, Ge Y, Xie H, Shi J, Deng L, Wu S, Shi H. Predicted lean body mass trajectories, and cancer risk and cancer-specific and all-cause mortality: a prospective cohort study. J Cachexia Sarcopenia Muscle 14: 2916–2924, 2023. doi:10.1002/jcsm.13370.
    Crossref | PubMed | Web of Science | Google Scholar
  • 691. Westbury LD, Beaudart C, Bruyère O, Cauley JA, Cawthon P, Cruz-Jentoft AJ, Curtis EM, Ensrud K, Fielding RA, Johansson H, Kanis JA, Karlsson MK, Lane NE, Lengelé L, Lorentzon M, McCloskey E, Mellström D, Newman AB, Ohlsson C, Orwoll E, Reginster JY, Ribom E, Rosengren BE, Schousboe JT, Shiroma EJ, Harvey NC, Dennison EM, Cooper C; International Musculoskeletal Ageing Network. Recent sarcopenia definitions-prevalence, agreement and mortality associations among men: findings from population-based cohorts. J Cachexia Sarcopenia Muscle 14: 565–575, 2023. doi:10.1002/jcsm.13160.
    Crossref | PubMed | Web of Science | Google Scholar
  • 692. Liu B, Liu R, Jin Y, Ding Y, Luo C. Association between possible sarcopenia, all-cause mortality, and adverse health outcomes in community-dwelling older adults in China. Sci Rep 14: 25913, 2024. doi:10.1038/s41598-024-77725-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 693. Zhang Y, Zhang J, Zhan Y, Pan Z, Liu Q, Yuan W. Sarcopenia is a prognostic factor of adverse effects and mortality in patients with tumour: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 15: 2295–2310, 2024. doi:10.1002/jcsm.13629.
    Crossref | PubMed | Web of Science | Google Scholar
  • 694. Lemos T, Gallagher D. Current body composition measurement techniques. Curr Opin Endocrinol Diabetes Obes 24: 310–314, 2017. doi:10.1097/MED.0000000000000360.
    Crossref | PubMed | Web of Science | Google Scholar
  • 695. Hemke R, Buckless C, Torriani M. Quantitative imaging of body composition. Semin Musculoskelet Radiol 24: 375–385, 2020. doi:10.1055/s-0040-1708824.
    Crossref | PubMed | Web of Science | Google Scholar
  • 696. Duren DL, Sherwood RJ, Czerwinski SA, Lee M, Choh AC, Siervogel RM, Cameron Chumlea W. Body composition methods: comparisons and interpretation. J Diabetes Sci Technol 2: 1139–1146, 2008. doi:10.1177/193229680800200623.
    Crossref | PubMed | Google Scholar
  • 697. Sales WB, Mâcedo S, Gonçalves R, Andrade LE, Ramalho CS, de Souza GF, Maciel AC. Use of electrical bioimpedance in the assessment of sarcopenia in the older adults: a scoping review. J Bodyw Mov Ther 39: 373–381, 2024. doi:10.1016/j.jbmt.2024.02.015.
    Crossref | PubMed | Web of Science | Google Scholar
  • 698. Börgeson E, Tavajoh S, Lange S, Jessen N. The challenges of assessing adiposity in a clinical setting. Nat Rev Endocrinol 20: 615–626, 2024. doi:10.1038/s41574-024-01012-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 699. Mijnarends DM, Meijers JM, Halfens RJ, ter Borg S, Luiking YC, Verlaan S, Schoberer D, Cruz Jentoft AJ, van Loon LJ, Schols JM. Validity and reliability of tools to measure muscle mass, strength, and physical performance in community-dwelling older people: a systematic review. J Am Med Dir Assoc 14: 170–178, 2013. doi:10.1016/j.jamda.2012.10.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 700. Sabatino A, Sola KH, Brismar TB, Lindholm B, Stenvinkel P, Avesani CM. Making the invisible visible: imaging techniques for assessing muscle mass and muscle quality in chronic kidney disease. Clin Kidney J 17: sfae028, 2024. doi:10.1093/ckj/sfae028.
    Crossref | PubMed | Web of Science | Google Scholar
  • 701. Lukaski H, Raymond-Pope CJ. New frontiers of body composition in sport. Int J Sports Med 42: 588–601, 2021. doi:10.1055/a-1373-5881.
    Crossref | PubMed | Web of Science | Google Scholar
  • 702. Golja P, Robič Pikel T, Zdešar Kotnik K, Fležar M, Selak S, Kapus J, Kotnik P. Direct comparison of (anthropometric) methods for the assessment of body composition. Ann Nutr Metab 76: 183–192, 2020. doi:10.1159/000508514.
    Crossref | PubMed | Web of Science | Google Scholar
  • 703. Heymsfield SB. Advances in body composition: a 100-year journey. Int J Obes (Lond) 49: 177–181, 2025. doi:10.1038/s41366-024-01511-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 704. Nana A, Slater GJ, Stewart AD, Burke LM. Methodology review: using dual-energy X-ray absorptiometry (DXA) for the assessment of body composition in athletes and active people. Int J Sport Nutr Exerc Metab 25: 198–215, 2015. doi:10.1123/ijsnem.2013-0228.
    Crossref | PubMed | Web of Science | Google Scholar
  • 705. Oliver CJ, Climstein M, Rosic N, Bosy-Westphal A, Tinsley G, Myers S. Fat-free mass: friend or foe to metabolic health? J Cachexia Sarcopenia Muscle 16: e13714, 2025. doi:10.1002/jcsm.13714.
    Crossref | PubMed | Web of Science | Google Scholar
  • 706. Rodriguez C, Mota JD, Palmer TB, Heymsfield SB, Tinsley GM. Skeletal muscle estimation: a review of techniques and their applications. Clin Physiol Funct Imaging 44: 261–284, 2024. doi:10.1111/cpf.12874.
    Crossref | PubMed | Web of Science | Google Scholar
  • 707. Seabolt LA, Welch EB, Silver HJ. Imaging methods for analyzing body composition in human obesity and cardiometabolic disease. Ann NY Acad Sci 1353: 41–59, 2015. doi:10.1111/nyas.12842.
    Crossref | PubMed | Web of Science | Google Scholar
  • 708. Fosbøl MO, Zerahn B. Contemporary methods of body composition measurement. Clin Physiol Funct Imaging 35: 81–97, 2015. doi:10.1111/cpf.12152.
    Crossref | PubMed | Web of Science | Google Scholar
  • 709. Lavalle S, Valerio MR, Masiello E, Gebbia V, Scandurra G. Unveiling the intricate dance: how cancer orchestrates muscle wasting and sarcopenia. In Vivo 38: 1520–1529, 2024. doi:10.21873/invivo.13602.
    Crossref | PubMed | Web of Science | Google Scholar
  • 710. Prado CM, Heymsfield SB. Lean tissue imaging: a new era for nutritional assessment and intervention. JPEN J Parenter Enteral Nutr 38: 940–953, 2014. doi:10.1177/0148607114550189.
    Crossref | PubMed | Web of Science | Google Scholar
  • 711. Evans WJ, Ferrucci L. A simplified definition of sarcopenia: muscle mass/body weight. J Nutr Health Aging 28: 100302, 2024. doi:10.1016/j.jnha.2024.100302.
    Crossref | PubMed | Web of Science | Google Scholar
  • 712. Pagano AP, Montenegro J, Oliveira CL, Desai N, Gonzalez MC, Cawthon PM, Evans WJ, Prado CM. Estimating muscle mass using D3-creatine dilution: a narrative review of clinical implications and comparison with other methods. J Gerontol A Biol Sci Med Sci 79: glad280, 2024. doi:10.1093/gerona/glad280.
    Crossref | PubMed | Web of Science | Google Scholar
  • 713. McCarthy C, Schoeller D, Brown JC, Gonzalez MC, Varanoske AN, Cataldi D, Shepherd J, Heymsfield SB. D3-creatine dilution for skeletal muscle mass measurement: historical development and current status. J Cachexia Sarcopenia Muscle 13: 2595–2607, 2022. doi:10.1002/jcsm.13083.
    Crossref | PubMed | Web of Science | Google Scholar
  • 714. Cawthon PM, Blackwell T, Cummings SR, Orwoll ES, Duchowny KA, Kado DM, Stone KL, Ensrud KE, Cauley JA, Evans WJ. Muscle mass assessed by the D3-creatine dilution method and incident self-reported disability and mortality in a prospective observational study of community-dwelling older men. J Gerontol A Biol Sci Med Sci 76: 123–130, 2021. doi:10.1093/gerona/glaa111.
    Crossref | PubMed | Web of Science | Google Scholar
  • 715. Evans WJ, Hellerstein M, Orwoll E, Cummings S, Cawthon PM. D3-creatine dilution and the importance of accuracy in the assessment of skeletal muscle mass. J Cachexia Sarcopenia Muscle 10: 14–21, 2019. doi:10.1002/jcsm.12390.
    Crossref | PubMed | Web of Science | Google Scholar
  • 716. Fujita H. AI-based computer-aided diagnosis (AI-CAD): the latest review to read first. Radiol Phys Technol 13: 6–19, 2020. doi:10.1007/s12194-019-00552-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 717. Pooler BD, Garrett JW, Lee MH, Rush BE, Kuchnia AJ, Summers RM, Pickhardt PJ. CT-based body composition measures and systemic disease: a population-level analysis using artificial intelligence tools in over 100,000 patients. AJR Am J Roentgenol 224: e2432216, 2025. doi:10.2214/AJR.24.32216.
    Crossref | PubMed | Google Scholar
  • 718. de Borba EL, Ceolin J, Ziegelmann PK, Bodanese LC, Gonçalves MR, Cañon-Montañez W, Mattiello R. Phase angle of bioimpedance at 50 kHz is associated with cardiovascular diseases: systematic review and meta-analysis. Eur J Clin Nutr 76: 1366–1373, 2022. doi:10.1038/s41430-022-01131-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 719. Langer RD, Ward LC, Larsen SC, Heitmann BL. Can change in phase angle predict the risk of morbidity and mortality during an 18-year follow-up period? A cohort study among adults. Front Nutr 10: 1157531, 2023. doi:10.3389/fnut.2023.1157531.
    Crossref | PubMed | Web of Science | Google Scholar
  • 720. Ballarin G, Valerio G, Alicante P, Di Vincenzo O, Monfrecola F, Scalfi L. Could BIA-derived phase angle predict health-related musculoskeletal fitness? A cross-sectional study in young adults. Nutrition 122: 112388, 2024. doi:10.1016/j.nut.2024.112388.
    Crossref | PubMed | Web of Science | Google Scholar
  • 721. Ward LC, Brantlov S. Bioimpedance basics and phase angle fundamentals. Rev Endocr Metab Disord 24: 381–391, 2023. doi:10.1007/s11154-022-09780-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 722. Silva AM, Campa F, Stagi S, Gobbo LA, Buffa R, Toselli S, , et al.. The bioelectrical impedance analysis (BIA) international database: aims, scope, and call for data. Eur J Clin Nutr 77: 1143–1150, 2023. doi:10.1038/s41430-023-01310-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 723. Pratt J, Narici M, Boreham C, De Vito G. Dual-energy x-ray absorptiometry derived body composition trajectories across adulthood: reference values and associations with body roundness index and body mass index. Clin Nutr 46: 137–146, 2025. doi:10.1016/j.clnu.2025.02.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 724. Borda MG, Patricio Baldera J, Patino-Hernandez D, Westman E, Pérez-Zepeda MU, Tarazona-Santabalbina FJ, Wakabayashi H, Arai H, Kivipelto M, Aarsland D. Temporal muscle thickness predicts mortality and disability in older adults diagnosed with mild dementia. J Frailty Aging 13: 441–447, 2024. doi:10.14283/jfa.2024.39.
    Crossref | PubMed | Web of Science | Google Scholar
  • 725. Grune E, Nattenmüller J, Kiefer LS, Machann J, Peters A, Bamberg F, Schlett CL, Rospleszcz S. Subphenotypes of body composition and their association with cardiometabolic risk - Magnetic resonance imaging in a population-based sample. Metabolism 164: 156130, 2025. doi:10.1016/j.metabol.2024.156130.
    Crossref | PubMed | Web of Science | Google Scholar
  • 726. Garcia-Diez AI, Porta-Vilaro M, Isern-Kebschull J, Naude N, Guggenberger R, Brugnara L, Milinkovic A, Bartolome-Solanas A, Soler-Perromat JC, Del Amo M, Novials A, Tomas X. Myosteatosis: diagnostic significance and assessment by imaging approaches. Quant Imaging Med Surg 14: 7937–7957, 2024. doi:10.21037/qims-24-365.
    Crossref | PubMed | Web of Science | Google Scholar
  • 727. Souza AC, Troschel AS, Marquardt JP, Hadžić I, Foldyna B, Moura FA, Hainer J, Divakaran S, Blankstein R, Dorbala S, Di Carli MF, Aerts HJ, Lu MT, Fintelmann FJ, Taqueti VR. Skeletal muscle adiposity, coronary microvascular dysfunction, and adverse cardiovascular outcomes. Eur Heart J 46: 1112–1123, 2025. doi:10.1093/eurheartj/ehae827.
    Crossref | PubMed | Web of Science | Google Scholar
  • 728. Chait A, den Hartigh LJ. Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med 7: 22, 2020. doi:10.3389/fcvm.2020.00022.
    Crossref | PubMed | Web of Science | Google Scholar
  • 729. Liang Y, Ye X, Pan M, Chen Y, Yuan Y, Luo L. Impact of steatotic liver disease subtypes, sarcopenia, and fibrosis on all-cause and cause-specific mortality: a 15.7-year cohort study. BMC Gastroenterol 25: 75, 2025. doi:10.1186/s12876-025-03661-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 730. Anderson DB, Beach AJ, Chen L, Feng HJ, McKay MJ, Smith ZA, Weber KA 2nd, Wesselink EO, Elliott JM. What is normal age-related thigh muscle composition among 45- to 84-year-old adults from the UK Biobank study. Geroscience 47: 1175–1185, 2025. doi:10.1007/s11357-024-01304-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 731. Lu A, Than S, Beare R, La Hood A, Collyer TA, Srikanth V, Moran C. Interactions between muscle volume and body mass index on brain structure in the UK Biobank. Front Dement 3: 1456716, 2024. doi:10.3389/frdem.2024.1456716.
    Crossref | PubMed | Google Scholar
  • 732. Pischon T, Boeing H, Hoffmann K, Bergmann M, Schulze MB, Overvad K, , et al.. General and abdominal adiposity and risk of death in Europe. N Engl J Med 359: 2105–2120, 2008. doi:10.1056/NEJMoa0801891.
    Crossref | PubMed | Web of Science | Google Scholar
  • 733. Sánchez-Romero LM, Sagaceta-Mejía J, Mindell JS, Passi-Solar A, Bernabé-Ortiz A, Tolentino-Mayo L, Moody A, Scholes S. Sex differences in the secular change in waist circumference relative to BMI in five countries from 1997 to 2020. Obesity (Silver Spring) 32: 1934–1947, 2024. doi:10.1002/oby.24110.
    Crossref | PubMed | Web of Science | Google Scholar
  • 734. Tao Z, Zuo P, Ma G. The association between weight-adjusted waist circumference index and cardiovascular disease and mortality in patients with diabetes. Sci Rep 14: 18973, 2024. doi:10.1038/s41598-024-69712-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 735. Shen Q, Zhou T, Chen X, Umar HM, Yang X, Shen X. Evaluating the weight-adjusted waist index as a predictive tool for sarcopenia and mortality risk. Eat Weight Disord 30: 1, 2025. doi:10.1007/s40519-024-01712-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 736. Zhang X, Ma N, Lin Q, Chen K, Zheng F, Wu J, Dong X, Niu W. Body roundness index and all-cause mortality among US adults. JAMA Netw Open 7: e2415051, 2024. doi:10.1001/jamanetworkopen.2024.15051.
    Crossref | PubMed | Web of Science | Google Scholar
  • 737. Liu Y, He H, Qian K, Huang Y, Ao X, Shi X, Ruan B, Xue R, Fu X, Wang S. Evaluation of health associations with height-normalised abdominal body composition indices: a single-centre cross-sectional study. J Cachexia Sarcopenia Muscle 15: 2651–2659, 2024. doi:10.1002/jcsm.13609.
    Crossref | PubMed | Web of Science | Google Scholar
  • 738. Benz E, Pinel A, Guillet C, Capel F, Pereira B, De Antonio M, Pouget M, Cruz-Jentoft AJ, Eglseer D, Topinkova E, Barazzoni R, Rivadeneira F, Ikram MA, Steur M, Voortman T, Schoufour JD, Weijs PJ, Boirie Y. Sarcopenia and sarcopenic obesity and mortality among older people. JAMA Netw Open 7: e243604, 2024. doi:10.1001/jamanetworkopen.2024.3604.
    Crossref | PubMed | Web of Science | Google Scholar
  • 739. Gengxin Y, Xuehan M, Xinyu W, Yali Y, Yiran X, Lishuang Z, Yiming Q, Guichen L, Li C. Association between sarcopenic obesity and risk of frailty in older adults: a systematic review and meta-analysis. Age Ageing 54: afae286, 2025. doi:10.1093/ageing/afae286.
    Crossref | PubMed | Web of Science | Google Scholar
  • 740. Booranasuksakul U, Guan Z, Macdonald IA, Tsintzas K, Stephan BC, Siervo M. Sarcopenic obesity and brain health: a critical appraisal of the current evidence. Nutr Bull 50: 30–43, 2025. doi:10.1111/nbu.12725.
    Crossref | PubMed | Web of Science | Google Scholar
  • 741. Briand M, Raffin J, Gonzalez-Bautista E, Ritz P, Abellan Van Kan G, Pillard F, Faruch-Bilfeld M, Guyonnet S, Dray C, Vellas B, de Souto Barreto P, Rolland Y. Body composition and aging: cross-sectional results from the INSPIRE study in people 20 to 93 years old. Geroscience 47: 863–875, 2025. doi:10.1007/s11357-024-01245-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 742. Sedlmeier AM, Baumeister SE, Weber A, Fischer B, Thorand B, Ittermann T, Dörr M, Felix SB, Völzke H, Peters A, Leitzmann MF. Relation of body fat mass and fat-free mass to total mortality: results from 7 prospective cohort studies. Am J Clin Nutr 113: 639–646, 2021. doi:10.1093/ajcn/nqaa339.
    Crossref | PubMed | Web of Science | Google Scholar
  • 743. Damanti S, Citterio L, Zagato L, Brioni E, Magnaghi C, Simonini M, De Lorenzo R, Ruggiero M, Santoro S, Senini E, Messina M, Vitali G, Manunta P, Manfredi AA, Lanzani C, Querini PR. Sarcopenic obesity and pre-sarcopenia contribute to frailty in community-dwelling Italian older people: data from the FRASNET study. BMC Geriatr 24: 638, 2024. doi:10.1186/s12877-024-05216-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 744. Linge J, Ekstedt M, Dahlqvist Leinhard O. Adverse muscle composition is linked to poor functional performance and metabolic comorbidities in NAFLD. JHEP Rep 3: 100197, 2021. doi:10.1016/j.jhepr.2020.100197.
    Crossref | PubMed | Web of Science | Google Scholar
  • 745. Linge J, Nasr P, Sanyal AJ, Dahlqvist Leinhard O, Ekstedt M. Adverse muscle composition is a significant risk factor for all-cause mortality in NAFLD. JHEP Rep 5: 100663, 2023. doi:10.1016/j.jhepr.2022.100663.
    Crossref | PubMed | Web of Science | Google Scholar
  • 746. Arden NK, Spector TD. Genetic influences on muscle strength, lean body mass, and bone mineral density: a twin study. J Bone Miner Res 12: 2076–2081, 1997. doi:10.1359/jbmr.1997.12.12.2076.
    Crossref | PubMed | Web of Science | Google Scholar
  • 747. GBD 2021 Adult BMI Collaborators. Global, regional, and national prevalence of adult overweight and obesity, 1990–2021, with forecasts to 2050: a forecasting study for the Global Burden of Disease Study 2021. Lancet 405: 813–838, 2025. doi:10.1016/S0140-6736(25)00355-1.
    Crossref | PubMed | Google Scholar
  • 748. Cruz-Jentoft AJ, Gonzalez MC, Prado CM. Sarcopenia ≠ low muscle mass. Eur Geriatr Med 14: 225–228, 2023. doi:10.1007/s41999-023-00760-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 749. Manini TM, Clark BC. Dynapenia and aging: an update. J Gerontol A Biol Sci Med Sci 67: 28–40, 2012. doi:10.1093/gerona/glr010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 750. Freitas SR, Cruz-Montecinos C, Ratel S, Pinto RS. Powerpenia should be considered a biomarker of healthy aging. Sports Med Open 10: 27, 2024. doi:10.1186/s40798-024-00689-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 751. Clark BC, Manini TM. Sarcopenia ≠ dynapenia. J Gerontol A Biol Sci Med Sci 63: 829–834, 2008. doi:10.1093/gerona/63.8.829.
    Crossref | PubMed | Web of Science | Google Scholar
  • 752. Araújo CG, Tou NX. Muscle matters: bridging the gap between terminology of age-related muscle loss and exercise interventions. J Aging Phys Act 2024: 1–3, 2024. doi:10.1123/japa.2024-0111.
    Crossref | PubMed | Web of Science | Google Scholar
  • 753. Mitchell WK, Williams J, Atherton P, Larvin M, Lund J, Narici M. Sarcopenia, dynapenia, and the impact of advancing age on human skeletal muscle size and strength; a quantitative review. Front Physiol 3: 260, 2012. doi:10.3389/fphys.2012.00260.
    Crossref | PubMed | Web of Science | Google Scholar
  • 754. van den Hoek DJ, Beaumont PL, van den Hoek AK, Owen PJ, Garrett JM, Buhmann R, Latella C. Normative data for the squat, bench press and deadlift exercises in powerlifting: data from 809,986 competition entries. J Sci Med Sport 27: 734–742, 2024. doi:10.1016/j.jsams.2024.07.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 755. Wiegmann S, Felsenberg D, Armbrecht G, Dietzel R. Longitudinal changes in muscle power compared to muscle strength and mass. J Musculoskelet Neuronal Interact 21: 13–25, 2021. https://www.ismni.org/jmni/article/21/13.
    PubMed | Web of Science | Google Scholar
  • 756. Brown JC, Harhay MO, Harhay MN. The muscle quality index and mortality among males and females. Ann Epidemiol 26: 648–653, 2016. doi:10.1016/j.annepidem.2016.07.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 757. Fragala MS, Kenny AM, Kuchel GA. Muscle quality in aging: a multi-dimensional approach to muscle functioning with applications for treatment. Sports Med 45: 641–658, 2015. doi:10.1007/s40279-015-0305-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 758. McGregor RA, Cameron-Smith D, Poppitt SD. It is not just muscle mass: a review of muscle quality, composition and metabolism during ageing as determinants of muscle function and mobility in later life. Longev Healthspan 3: 9, 2014. doi:10.1186/2046-2395-3-9.
    Crossref | PubMed | Google Scholar
  • 759. Furrer R, Handschin C. Muscle wasting diseases: novel targets and treatments. Annu Rev Pharmacol Toxicol 59: 315–339, 2019. doi:10.1146/annurev-pharmtox-010818-021041.
    Crossref | PubMed | Web of Science | Google Scholar
  • 760. Kanbay M, Siriopol D, Copur S, Hasbal NB, Güldan M, Kalantar-Zadeh K, Garfias-Veitl T, von Haehling S. Effect of Bimagrumab on body composition: a systematic review and meta-analysis. Aging Clin Exp Res 36: 185, 2024. doi:10.1007/s40520-024-02825-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 761. Zhang S, Peng LN, Lee WJ, Nishita Y, Otsuka R, Arai H, Chen LK. Muscle function outweighs appendicular lean mass in predicting adverse outcomes: evidence from Asian longitudinal studies. J Nutr Health Aging 28: 100403, 2024. doi:10.1016/j.jnha.2024.100403.
    Crossref | PubMed | Web of Science | Google Scholar
  • 762. Li R, Xia J, Zhang XI, Gathirua-Mwangi WG, Guo J, Li Y, McKenzie S, Song Y. Associations of muscle mass and strength with all-cause mortality among US older adults. Med Sci Sports Exerc 50: 458–467, 2018. doi:10.1249/MSS.0000000000001448.
    Crossref | PubMed | Web of Science | Google Scholar
  • 763. Newman AB, Kupelian V, Visser M, Simonsick EM, Goodpaster BH, Kritchevsky SB, Tylavsky FA, Rubin SM, Harris TB. Strength, but not muscle mass, is associated with mortality in the health, aging and body composition study cohort. J Gerontol A Biol Sci Med Sci 61: 72–77, 2006. doi:10.1093/gerona/61.1.72.
    Crossref | PubMed | Web of Science | Google Scholar
  • 764. Sayer AA, Kirkwood TB. Grip strength and mortality: a biomarker of ageing? Lancet 386: 226–227, 2015. doi:10.1016/S0140-6736(14)62349-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 765. Soysal P, Hurst C, Demurtas J, Firth J, Howden R, Yang L, Tully MA, Koyanagi A, Ilie PC, López-Sánchez GF, Schwingshackl L, Veronese N, Smith L. Handgrip strength and health outcomes: umbrella review of systematic reviews with meta-analyses of observational studies. J Sport Health Sci 10: 290–295, 2021. doi:10.1016/j.jshs.2020.06.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 766. Sáez de Asteasu ML, Cadore EL, Steffens T, Blanco-Rambo E, Schneider TC, Izquierdo M, Pietta-Dias C. Reduced handgrip strength is associated with 1 year-mortality in Brazilian frail nonagenarians and centenarians. J Frailty Aging 13: 31–34, 2024. doi:10.14283/jfa.2023.21.
    Crossref | PubMed | Web of Science | Google Scholar
  • 767. García-Hermoso A, Cavero-Redondo I, Ramírez-Vélez R, Ruiz JR, Ortega FB, Lee DC, Martínez-Vizcaíno V. Muscular strength as a predictor of all-cause mortality in an apparently healthy population: a systematic review and meta-analysis of data from approximately 2 million men and women. Arch Phys Med Rehabil 99: 2100–2113.e5, 2018. doi:10.1016/j.apmr.2018.01.008.
    Crossref | PubMed | Web of Science | Google Scholar
  • 768. Vaishya R, Misra A, Vaish A, Ursino N, D'Ambrosi R. Hand grip strength as a proposed new vital sign of health: a narrative review of evidences. J Health Popul Nutr 43: 7, 2024. doi:10.1186/s41043-024-00500-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 769. Faigenbaum AD, Garcia-Hermoso A, MacDonald JP, Mortatti A, Rial Rebullido T. Bridging the gap between strengthspan and lifespan. Br J Sports Med 58: 758–760, 2024. doi:10.1136/bjsports-2024-108357.
    Crossref | PubMed | Web of Science | Google Scholar
  • 770. Volaklis KA, Halle M, Meisinger C. Muscular strength as a strong predictor of mortality: a narrative review. Eur J Intern Med 26: 303–310, 2015. doi:10.1016/j.ejim.2015.04.013.
    Crossref | PubMed | Web of Science | Google Scholar
  • 771. Jinha A, Herzog W. Muscle power: a simple concept causing much confusion. J Sport Health Sci 14: 101005, 2024. doi:10.1016/j.jshs.2024.101005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 772. Coelho-Junior HJ, Álvarez-Bustos A, Landi F, da Silva Aguiar S, Rodriguez-Mañas L, Marzetti E. Why are we not exploring the potential of lower limb muscle power to identify people with sarcopenia? Ageing Res Rev 104: 102662, 2025. doi:10.1016/j.arr.2025.102662.
    Crossref | PubMed | Web of Science | Google Scholar
  • 773. Alcazar J, Rodriguez-Lopez C, Delecluse C, Thomis M, Van Roie E. Ten-year longitudinal changes in muscle power, force, and velocity in young, middle-aged, and older adults. J Cachexia Sarcopenia Muscle 14: 1019–1032, 2023. doi:10.1002/jcsm.13184.
    Crossref | PubMed | Web of Science | Google Scholar
  • 774. Losa-Reyna J, Alcazar J, Carnicero J, Alfaro-Acha A, Castillo-Gallego C, Rosado-Artalejo C, Rodríguez-Mañas L, Ara I, Garcia-Garcia FJ. Impact of relative muscle power on hospitalization and all-cause mortality in older adults. J Gerontol A Biol Sci Med Sci 77: 781–789, 2022. doi:10.1093/gerona/glab230.
    Crossref | PubMed | Web of Science | Google Scholar
  • 775. Izquierdo M, Cadore EL. Power to prolong independence and healthy ageing in older adults. Br J Sports Med 58: 524–526, 2024. doi:10.1136/bjsports-2023-107683.
    Crossref | PubMed | Web of Science | Google Scholar
  • 776. McGrath R, McGrath BM, Jurivich D, Knutson P, Mastrud M, Singh B, Tomkinson GR. Collective weakness is associated with time to mortality in Americans. J Strength Cond Res 38: e398–e404, 2024. doi:10.1519/JSC.0000000000004780.
    Crossref | PubMed | Web of Science | Google Scholar
  • 777. Chan OY, van Houwelingen AH, Gussekloo J, Blom JW, den Elzen WP. Comparison of quadriceps strength and handgrip strength in their association with health outcomes in older adults in primary care. Age (Dordr) 36: 9714, 2014. doi:10.1007/s11357-014-9714-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 778. Guadalupe-Grau A, Carnicero JA, Gómez-Cabello A, Gutiérrez Avila G, Humanes S, Alegre LM, Castro M, Rodríguez-Mañas L, García-García FJ. Association of regional muscle strength with mortality and hospitalisation in older people. Age Ageing 44: 790–795, 2015. doi:10.1093/ageing/afv080.
    Crossref | PubMed | Web of Science | Google Scholar
  • 779. Andersen LL, López-Bueno R, Núñez-Cortés R, Cadore EL, Polo-López A, Calatayud J. Association of muscle strength with all-cause mortality in the oldest old: prospective cohort study from 28 countries. J Cachexia Sarcopenia Muscle 15: 2756–2764, 2024. doi:10.1002/jcsm.13619.
    Crossref | PubMed | Web of Science | Google Scholar
  • 780. FitzGerald SJ, Barlow CE, Kampert JB, Morrow JR, Jackson AW, Blair SN. Muscular fitness and all-cause mortality: prospective observations. J Phys Act Health 1: 7–18, 2004. doi:10.1123/jpah.1.1.7.
    Crossref | Google Scholar
  • 781. Andersen LL, Calatayud J, Núñez-Cortés R, Polo-López A, López-Bueno R. Graded association of muscle strength with all-cause and cause-specific mortality in older adults with diabetes: prospective cohort study across 28 countries. Diabetes Obes Metab 27: 312–319, 2025. doi:10.1111/dom.16019.
    Crossref | PubMed | Web of Science | Google Scholar
  • 782. Dodds RM, Syddall HE, Cooper R, Benzeval M, Deary IJ, Dennison EM, Der G, Gale CR, Inskip HM, Jagger C, Kirkwood TB, Lawlor DA, Robinson SM, Starr JM, Steptoe A, Tilling K, Kuh D, Cooper C, Sayer AA. Grip strength across the life course: normative data from twelve British studies. PLoS One 9: e113637, 2014. doi:10.1371/journal.pone.0113637.
    Crossref | PubMed | Web of Science | Google Scholar
  • 783. Esteban-Cornejo I, Ho FK, Petermann-Rocha F, Lyall DM, Martinez-Gomez D, Cabanas-Sánchez V, Ortega FB, Hillman CH, Gill JM, Quinn TJ, Sattar N, Pell JP, Gray SR, Celis-Morales C. Handgrip strength and all-cause dementia incidence and mortality: findings from the UK Biobank prospective cohort study. J Cachexia Sarcopenia Muscle 13: 1514–1525, 2022. doi:10.1002/jcsm.12857.
    Crossref | PubMed | Web of Science | Google Scholar
  • 784. Leong DP, Teo KK, Rangarajan S, Lopez-Jaramillo P, Avezum A Jr, Orlandini A, Seron P, Ahmed SH, Rosengren A, Kelishadi R, Rahman O, Swaminathan S, Iqbal R, Gupta R, Lear SA, Oguz A, Yusoff K, Zatonska K, Chifamba J, Igumbor E, Mohan V, Anjana RM, Gu H, Li W, Yusuf S; Prospective Urban Rural Epidemiology Study investigators. Prognostic value of grip strength: findings from the Prospective Urban Rural Epidemiology (PURE) study. Lancet 386: 266–273, 2015. doi:10.1016/S0140-6736(14)62000-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 785. Roberts HC, Denison HJ, Martin HJ, Patel HP, Syddall H, Cooper C, Sayer AA. A review of the measurement of grip strength in clinical and epidemiological studies: towards a standardised approach. Age Ageing 40: 423–429, 2011. doi:10.1093/ageing/afr051.
    Crossref | PubMed | Web of Science | Google Scholar
  • 786. Günther CM, Bürger A, Rickert M, Crispin A, Schulz CU. Grip strength in healthy caucasian adults: reference values. J Hand Surg Am 33: 558–565, 2008. doi:10.1016/j.jhsa.2008.01.008.
    Crossref | PubMed | Web of Science | Google Scholar
  • 787. Steiber N. Strong or weak handgrip? Normative reference values for the German population across the life course stratified by sex, age, and body height. PLoS One 11: e0163917, 2016. doi:10.1371/journal.pone.0163917.
    Crossref | PubMed | Web of Science | Google Scholar
  • 788. Wang YC, Bohannon RW, Li X, Sindhu B, Kapellusch J. Hand-grip strength: normative reference values and equations for individuals 18 to 85 years of age residing in the United States. J Orthop Sports Phys Ther 48: 685–693, 2018. doi:10.2519/jospt.2018.7851.
    Crossref | PubMed | Web of Science | Google Scholar
  • 789. Silva-Santos T, Guerra RS, Valdiviesso R, Amaral TF. Hand grip force-time curve indicators evaluated by dynamometer: a systematic review. Nutrients 16, 2024. doi:10.3390/nu16121951.
    Crossref | PubMed | Web of Science | Google Scholar
  • 790. Rijk JM, Roos PR, Deckx L, van den Akker M, Buntinx F. Prognostic value of handgrip strength in people aged 60 years and older: a systematic review and meta-analysis. Geriatr Gerontol Int 16: 5–20, 2016. doi:10.1111/ggi.12508.
    Crossref | PubMed | Web of Science | Google Scholar
  • 791. Peterson MD, Casten K, Collins S, Hassan H, García-Hermoso A, Faul J. Muscle weakness is a prognostic indicator of disability and chronic disease multimorbidity. Exp Gerontol 152: 111462, 2021. doi:10.1016/j.exger.2021.111462.
    Crossref | PubMed | Web of Science | Google Scholar
  • 792. Lee A, Park S. Factors affecting handgrip strength in menopausal women at high risk of sarcopenia: a national population-based study. Healthcare (Basel) 12, 2024. doi:10.3390/healthcare12242590.
    Crossref | PubMed | Web of Science | Google Scholar
  • 793. Syddall H, Cooper C, Martin F, Briggs R, Aihie Sayer A. Is grip strength a useful single marker of frailty? Age Ageing 32: 650–656, 2003. doi:10.1093/ageing/afg111.
    Crossref | PubMed | Web of Science | Google Scholar
  • 794. Granic A, Cooper R, Hurst C, Hillman SJ, Dodds RM, Witham MD, Sayer AA. Cross-sectional and longitudinal associations between glycaemic measures and grip strength in people without diabetes in the UK Biobank cohort study. Eur Geriatr Med 16: 67–77, 2025. doi:10.1007/s41999-024-01119-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 795. Toba A. Combining body mass index and muscle function in investigating mortality among older hypertensive patients. Hypertens Res 48: 1201–1203, 2025. doi:10.1038/s41440-024-02032-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 796. Carson RG. Get a grip: individual variations in grip strength are a marker of brain health. Neurobiol Aging 71: 189–222, 2018. doi:10.1016/j.neurobiolaging.2018.07.023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 797. Zhang F, Luo B, Bai Y, Zhang Y, Huang L, Lu W. Association of handgrip strength and risk of cardiovascular disease: a population-based cohort study. Aging Clin Exp Res 36: 207, 2024. doi:10.1007/s40520-024-02856-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 798. Amarowicz J, Warzecha M, Krawczyk A. Handgrip and its relation to age, fragility fractures, and BMD between sexes in a population aged 50+ years. Prz Menopauzalny 23: 167–172, 2024. doi:10.5114/pm.2024.145947.
    Crossref | PubMed | Web of Science | Google Scholar
  • 799. Kaczorowska A, Kozieł S, Ignasiak Z. Hand grip strength and quality of life among adults aged 50-90 years from South West Poland. Sci Rep 15: 882, 2025. doi:10.1038/s41598-024-84923-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 800. Ribeiro LW, Berndt S, Mielke GI, Doust J, Mishra GD. Factors associated with handgrip strength across the life course: a systematic review. J Cachexia Sarcopenia Muscle 15: 2270–2280, 2024. doi:10.1002/jcsm.13586.
    Crossref | PubMed | Web of Science | Google Scholar
  • 801. Dos Santos AP, Cordeiro JF, Abdalla PP, Bohn L, Sebastiño E, da Silva LS, Tasinafo-Júnior MF, Venturini AC, Andaki AC, Mendes EL, Marcos-Pardo PJ, Mota J, Machado DR. Low handgrip strength is associated with falls after the age of 50: findings from the Brazilian longitudinal study of aging (ELSI-Brazil). Arch Public Health 82: 172, 2024. doi:10.1186/s13690-024-01340-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 802. Tanaka T, Lyu W, Yoshizawa Y, Son BK, Iijima K. Letter to the Editor: “kami-chigiri” (newspaper tear-off) test: simple screening method for assessing muscle weakness among community-dwelling older adults. J Frailty Aging 13: 586–587, 2024. doi:10.14283/jfa.2024.62.
    Crossref | PubMed | Web of Science | Google Scholar
  • 803. Yeung SS, Reijnierse EM, Trappenburg MC, Hogrel JY, McPhee JS, Piasecki M, Sipila S, Salpakoski A, Butler-Browne G, Päaäsuke M, Gapeyeva H, Narici MV, Meskers CG, Maier AB. Handgrip strength cannot be assumed a proxy for overall muscle strength. J Am Med Dir Assoc 19: 703–709, 2018. doi:10.1016/j.jamda.2018.04.019.
    Crossref | PubMed | Web of Science | Google Scholar
  • 804. Swales B, Ryde GC, Fletcher I, Whittaker AC. The reliability and suitability of strength assessments in frail and pre-frail older adults: recommendations for strength testing in older populations. BMC Geriatr 23: 820, 2023. doi:10.1186/s12877-023-04552-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 805. Arnold WD, Clark BC. Neuromuscular junction transmission failure in aging and sarcopenia: the nexus of the neurological and muscular systems. Ageing Res Rev 89: 101966, 2023. doi:10.1016/j.arr.2023.101966.
    Crossref | PubMed | Web of Science | Google Scholar
  • 806. Verschueren A, Palminha C, Delmont E, Attarian S. Changes in neuromuscular function in elders: novel techniques for assessment of motor unit loss and motor unit remodeling with aging. Rev Neurol (Paris) 178: 780–787, 2022. doi:10.1016/j.neurol.2022.03.019.
    Crossref | PubMed | Web of Science | Google Scholar
  • 807. Virto N, Río X, Méndez-Zorrilla A, García-Zapirain B. Non invasive techniques for direct muscle quality assessment after exercise intervention in older adults: a systematic review. BMC Geriatr 24: 642, 2024. doi:10.1186/s12877-024-05243-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 808. Wang H, Zuo S, Cerezo-Sánchez M, Arekhloo NG, Nazarpour K, Heidari H. Wearable super-resolution muscle-machine interfacing. Front Neurosci 16: 1020546, 2022. doi:10.3389/fnins.2022.1020546.
    Crossref | PubMed | Web of Science | Google Scholar
  • 809. Olpe T, Wunderle C, Bargetzi L, Tribolet P, Laviano A, Stanga Z, Prado CM, Mueller B, Schuetz P. Muscle matters: prognostic implications of malnutrition and muscle health parameters in patients with cancer. A secondary analysis of a randomised trial. Clin Nutr 43: 2255–2262, 2024. doi:10.1016/j.clnu.2024.07.020.
    Crossref | PubMed | Web of Science | Google Scholar
  • 810. Joyner MJ, Wiggins CC, Baker SE, Klassen SA, Senefeld JW. Exercise and experiments of nature. Compr Physiol 13: 4879–4907, 2023. doi:10.1002/cphy.c220027.
    Crossref | PubMed | Web of Science | Google Scholar
  • 811. Ding D, Ekelund U. From London buses to activity trackers: a reflection of 70 years of physical activity research. J Sport Health Sci 13: 736–738, 2024. doi:10.1016/j.jshs.2024.06.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 812. Morris JN, Heady JA, Raffle PA, Roberts CG, Parks JW. Coronary heart-disease and physical activity of work. Lancet 262: 1111–1120, 1953. doi:10.1016/s0140-6736(53)91495-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 813. Morris JN, Heady JA, Raffle PA, Roberts CG, Parks JW. Coronary heart-disease and physical activity of work. Lancet 262: 1053–1057, 1953. doi:10.1016/s0140-6736(53)90665-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 814. Gremaud AL, Carr LJ, Simmering JE, Evans NJ, Cremer JF, Segre AM, Polgreen LA, Polgreen PM. Gamifying accelerometer use increases physical activity levels of sedentary office workers. J Am Heart Assoc 7: e007735, 2018. doi:10.1161/JAHA.117.007735.
    Crossref | PubMed | Web of Science | Google Scholar
  • 815. Piercy KL, Troiano RP, Ballard RM, Carlson SA, Fulton JE, Galuska DA, George SM, Olson RD. The Physical Activity Guidelines for Americans. JAMA 320: 2020–2028, 2018. doi:10.1001/jama.2018.14854.
    Crossref | PubMed | Web of Science | Google Scholar
  • 816. Bull FC, Al-Ansari SS, Biddle S, Borodulin K, Buman MP, Cardon G, Carty C, Chaput JP, Chastin S, Chou R, Dempsey PC, DiPietro L, Ekelund U, Firth J, Friedenreich CM, Garcia L, Gichu M, Jago R, Katzmarzyk PT, Lambert E, Leitzmann M, Milton K, Ortega FB, Ranasinghe C, Stamatakis E, Tiedemann A, Troiano RP, van der Ploeg HP, Wari V, Willumsen JF. World Health Organization 2020 guidelines on physical activity and sedentary behaviour. Br J Sports Med 54: 1451–1462, 2020. doi:10.1136/bjsports-2020-102955.
    Crossref | PubMed | Web of Science | Google Scholar
  • 817. Du Y, Liu B, Sun Y, Snetselaar LG, Wallace RB, Bao W. Trends in adherence to the Physical Activity Guidelines for Americans for aerobic activity and time spent on sedentary behavior among US adults, 2007 to 2016. JAMA Netw Open 2: e197597, 2019. doi:10.1001/jamanetworkopen.2019.7597.
    Crossref | PubMed | Web of Science | Google Scholar
  • 818. Strain T, Flaxman S, Guthold R, Semenova E, Cowan M, Riley LM, Bull FC, Stevens GA; Country Data Author Group. National, regional, and global trends in insufficient physical activity among adults from 2000 to 2022: a pooled analysis of 507 population-based surveys with 5.7 million participants. Lancet Glob Health 12: e1232–e1243, 2024. doi:10.1016/S2214-109X(24)00150-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 819. Hallal PC, Andersen LB, Bull FC, Guthold R, Haskell W, Ekelund U; Lancet Physical Activity Series Working Group. Global physical activity levels: surveillance progress, pitfalls, and prospects. Lancet 380: 247–257, 2012. doi:10.1016/S0140-6736(12)60646-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 820. Guthold R, Stevens GA, Riley LM, Bull FC. Worldwide trends in insufficient physical activity from 2001 to 2016: a pooled analysis of 358 population-based surveys with 1.9 million participants. Lancet Glob Health 6: e1077–e1086, 2018. doi:10.1016/S2214-109X(18)30357-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 821. Ren Z, Zhang Y, Drenowatz C, Eather N, Hong J, Wang L, Yan J, Chen S. How many adults have sufficient muscle-strengthening exercise and the associated factors: a systematic review consisting of 2,629,508 participants. J Exerc Sci Fit 22: 359–368, 2024. doi:10.1016/j.jesf.2024.06.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 822. Abildso CG, Daily SM, Umstattd Meyer MR, Perry CK, Eyler A. Prevalence of meeting aerobic, muscle-strengthening, and combined physical activity guidelines during leisure time among adults, by rural-urban classification and region—United States, 2020. MMWR Morb Mortal Wkly Rep 72: 85–89, 2023. doi:10.15585/mmwr.mm7204a1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 823. Martinez-Gomez D, Rodriguez-Artalejo F, Ding D, Ekelund U, Cabanas-Sanchez V. Trends in the association between meeting the physical activity guidelines and risk of mortality in US adults. Prog Cardiovasc Dis 83: 116–123, 2024. doi:10.1016/j.pcad.2024.02.011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 824. Garber CE, Blissmer B, Deschenes MR, Franklin BA, Lamonte MJ, Lee IM, Nieman DC, Swain DP; American College of Sports Medicine. American College of Sports Medicine position stand. Quantity and quality of exercise for developing and maintaining cardiorespiratory, musculoskeletal, and neuromotor fitness in apparently healthy adults: guidance for prescribing exercise. Med Sci Sports Exerc 43: 1334–1359, 2011. doi:10.1249/MSS.0b013e318213fefb.
    Crossref | PubMed | Web of Science | Google Scholar
  • 825. Leitzmann MF, Park Y, Blair A, Ballard-Barbash R, Mouw T, Hollenbeck AR, Schatzkin A. Physical activity recommendations and decreased risk of mortality. Arch Intern Med 167: 2453–2460, 2007. doi:10.1001/archinte.167.22.2453.
    Crossref | PubMed | Web of Science | Google Scholar
  • 826. Zhao M, Veeranki SP, Magnussen CG, Xi B. Recommended physical activity and all cause and cause specific mortality in US adults: prospective cohort study. BMJ 370: m2031, 2020. doi:10.1136/bmj.m2031.
    Crossref | PubMed | Web of Science | Google Scholar
  • 827. Thornton JS, Morley WN, Sinha SK. Move more, age well: prescribing physical activity for older adults. CMAJ 197: E59–E67, 2025. doi:10.1503/cmaj.231336.
    Crossref | PubMed | Web of Science | Google Scholar
  • 828. Williams PT. Physical fitness and activity as separate heart disease risk factors: a meta-analysis. Med Sci Sports Exerc 33: 754–761, 2001. doi:10.1097/00005768-200105000-00012.
    Crossref | PubMed | Web of Science | Google Scholar
  • 829. Myers J, Kaykha A, George S, Abella J, Zaheer N, Lear S, Yamazaki T, Froelicher V. Fitness versus physical activity patterns in predicting mortality in men. Am J Med 117: 912–918, 2004. doi:10.1016/j.amjmed.2004.06.047.
    Crossref | PubMed | Web of Science | Google Scholar
  • 830. Ji Y, Atakan MM, Yan X, Wu J, Kuang J, Peng L. Reallocating just 10 min to moderate-to-vigorous physical activity from other components of 24-hour movement behaviors improves cardiovascular health in adults. BMC Public Health 24: 1768, 2024. doi:10.1186/s12889-024-19255-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 831. Sáez de Asteasu ML, Martínez-Velilla N, Zambom-Ferraresi F, Galbete A, Ramírez-Vélez R, Cadore EL, Abizanda P, Gómez-Pavón J, Izquierdo M. Dose-response relationship between exercise duration and enhanced function and cognition in acutely hospitalized older adults: a secondary analysis of a randomized clinical trial. Innov Aging 8: igae053, 2024. doi:10.1093/geroni/igae053.
    Crossref | PubMed | Web of Science | Google Scholar
  • 832. Su J, Jiang Y, Fan X, Tao R, Wu M, Lu Y, Hua Y, Jin J, Guo Y, Lv J, Pei P, Chen Z, Li L, Zhou J. Association between physical activity and cancer risk among Chinese adults: a 10-year prospective study. Int J Behav Nutr Phys Act 19: 150, 2022. doi:10.1186/s12966-022-01390-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 833. Merchant RA, Aprahamian I, Woo J, Vellas B, Morley JE. Editorial: Resilience and successful aging. J Nutr Health Aging 26: 652–656, 2022. doi:10.1007/s12603-022-1818-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 834. Cesari M, Araujo de Carvalho I, Amuthavalli Thiyagarajan J, Cooper C, Martin FC, Reginster JY, Vellas B, Beard JR. Evidence for the domains supporting the construct of intrinsic capacity. J Gerontol A Biol Sci Med Sci 73: 1653–1660, 2018. doi:10.1093/gerona/gly011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 835. Beard JR, Si Y, Liu Z, Chenoweth L, Hanewald K. Intrinsic capacity: validation of a new WHO concept for healthy aging in a longitudinal Chinese study. J Gerontol A Biol Sci Med Sci 77: 94–100, 2022. doi:10.1093/gerona/glab226.
    Crossref | PubMed | Web of Science | Google Scholar
  • 836. Veronese N, Honvo G, Amuthavalli Thiyagarajan J, Rizzoli R, Cooper C, Bruyère O, Mikton C, Sumi Y, Diaz T, Reginster JY, Banerjee A; members of the WHO Collaborating Center Working Group on Locomotor Capacity. Attributes and definitions of locomotor capacity in older people: a World Health Organisation (WHO) locomotor capacity working group meeting report. Aging Clin Exp Res 34: 481–483, 2022. doi:10.1007/s40520-022-02080-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 837. Dhakal A, Bobrin BD. Cognitive deficits. In: StatPearls. StatPearls Publishing, 2025. https://www.ncbi.nlm.nih.gov/books/NBK559052/.
    Google Scholar
  • 838. Bautmans I, Knoop V, Amuthavalli Thiyagarajan J, Maier AB, Beard JR, Freiberger E, Belsky D, Aubertin-Leheudre M, Mikton C, Cesari M, Sumi Y, Diaz T, Banerjee A; WHO Working Group on Vitality Capacity. WHO working definition of vitality capacity for healthy longevity monitoring. Lancet Healthy Longev 3: e789–e796, 2022. doi:10.1016/S2666-7568(22)00200-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 839. Jones R, Taylor TL, Mankowski RT, Dodds F, Hankes M, Hobson J, Lin Y, Saffold K, Sint Jago SC, Tharpe MA, Zumbro EL, Thalacker-Mercer A, Buford TW. Exercise training to preserve vitality capacity in ageing. Exp Physiol, 2024. doi:10.1113/EP091731.
    Crossref | PubMed | Web of Science | Google Scholar
  • 840. Wenger HA, Bell GJ. The interactions of intensity, frequency and duration of exercise training in altering cardiorespiratory fitness. Sports Med 3: 346–356, 1986. doi:10.2165/00007256-198603050-00004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 841. Wasfy MM, Lee IM. Examining the dose-response relationship between physical activity and health outcomes. NEJM Evid 1: EVIDra2200190, 2022. doi:10.1056/EVIDra2200190.
    Crossref | PubMed | Google Scholar
  • 842. Wasfy MM, Baggish AL. Exercise dose in clinical practice. Circulation 133: 2297–2313, 2016. doi:10.1161/CIRCULATIONAHA.116.018093.
    Crossref | PubMed | Web of Science | Google Scholar
  • 843. Ekelund U, Dalene KE, Tarp J, Lee IM. Physical activity and mortality: what is the dose response and how big is the effect? Br J Sports Med 54: 1125–1126, 2020. doi:10.1136/bjsports-2019-101765.
    Crossref | PubMed | Web of Science | Google Scholar
  • 844. Garcia L, Pearce M, Abbas A, Mok A, Strain T, Ali S, Crippa A, Dempsey PC, Golubic R, Kelly P, Laird Y, McNamara E, Moore S, de Sa TH, Smith AD, Wijndaele K, Woodcock J, Brage S. Non-occupational physical activity and risk of cardiovascular disease, cancer and mortality outcomes: a dose-response meta-analysis of large prospective studies. Br J Sports Med 57: 979–989, 2023. doi:10.1136/bjsports-2022-105669.
    Crossref | PubMed | Web of Science | Google Scholar
  • 845. Izquierdo-Gomez R, Martínez-Gomez D, Shields N, Del Rosario Ortola-Vidal M, Rodríguez-Artalejo F, Cabanas-Sánchez V. The role of physical activity in the association between disability and mortality among US older adults: a nationwide prospective cohort study. Geroscience 46: 3275–3285, 2024. doi:10.1007/s11357-024-01072-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 846. Moore AZ, Simonsick EM, Landman B, Schrack J, Wanigatunga AA, Ferrucci L. Correlates of life course physical activity in participants of the Baltimore longitudinal study of aging. Aging Cell 23: e14078, 2024. doi:10.1111/acel.14078.
    Crossref | PubMed | Web of Science | Google Scholar
  • 847. Webber BJ, Piercy KL, Hyde ET, Whitfield GP. Association of muscle-strengthening and aerobic physical activity with mortality in US adults aged 65 years or older. JAMA Netw Open 5: e2236778, 2022. doi:10.1001/jamanetworkopen.2022.36778.
    Crossref | PubMed | Web of Science | Google Scholar
  • 848. Ma T, Sirard J, Yang L, Li Y, Tsang S, Fu A. Revisiting the concept of bout: associations of moderate-to-vigorous physical activity sessions and non-sessions with mortality. Int J Behav Nutr Phys Act 21: 81, 2024. doi:10.1186/s12966-024-01631-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 849. Tarp J, Dalene KE, Fagerland MW, Steene-Johannesen J, Hansen BH, Anderssen SA, Hagströmer M, Dohrn IM, Dempsey PC, Wijndaele K, Brage S, Nordström A, Nordström P, Diaz KM, Howard VJ, Hooker SP, Morseth B, Hopstock LA, Sagelv EH, Yates T, Edwardson CL, Lee IM, Ekelund U. Physical activity volume, intensity, and mortality: harmonized meta-analysis of prospective cohort studies. Am J Prev Med 67: 887–896, 2024. doi:10.1016/j.amepre.2024.07.022.
    Crossref | PubMed | Web of Science | Google Scholar
  • 850. Manini TM, Everhart JE, Patel KV, Schoeller DA, Colbert LH, Visser M, Tylavsky F, Bauer DC, Goodpaster BH, Harris TB. Daily activity energy expenditure and mortality among older adults. JAMA 296: 171–179, 2006. doi:10.1001/jama.296.2.171.
    Crossref | PubMed | Web of Science | Google Scholar
  • 851. Ekelund U, Sanchez-Lastra MA, Dalene KE, Tarp J. Dose-response associations, physical activity intensity and mortality risk: a narrative review. J Sport Health Sci 13: 24–29, 2024. doi:10.1016/j.jshs.2023.09.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 852. Ahmadi MN, Clare PJ, Katzmarzyk PT, Del Pozo Cruz B, Lee IM, Stamatakis E. Vigorous physical activity, incident heart disease, and cancer: how little is enough? Eur Heart J 43: 4801–4814, 2022. doi:10.1093/eurheartj/ehac572.
    Crossref | PubMed | Web of Science | Google Scholar
  • 853. Cillekens B, Coenen P, Huysmans MA, Holtermann A, Troiano RP, Mork PJ, , et al.. Should workers be physically active after work? Associations of leisure-time physical activity with cardiovascular and all-cause mortality across occupational physical activity levels-An individual participant data meta-analysis. J Sport Health Sci 14: 100987, 2024. doi:10.1016/j.jshs.2024.100987.
    Crossref | PubMed | Web of Science | Google Scholar
  • 854. Coenen P, Huysmans MA, Holtermann A, Troiano RP, Mork PJ, Krokstad S, , et al.. Associations of occupational and leisure-time physical activity with all-cause mortality: an individual participant data meta-analysis. Br J Sports Med 58: 1527–1538, 2024. doi:10.1136/bjsports-2024-108117.
    Crossref | PubMed | Web of Science | Google Scholar
  • 855. Martinez-Gomez D, Cabanas-Sanchez V, Yu T, Rodriguez-Artalejo F, Ding D, Lee IM, Ekelund U. Long-term leisure-time physical activity and risk of all-cause and cardiovascular mortality: dose-response associations in a prospective cohort study of 210 327 Taiwanese adults. Br J Sports Med 56: 919–926, 2022. doi:10.1136/bjsports-2021-104961.
    Crossref | PubMed | Web of Science | Google Scholar
  • 856. Andersen LB, Schnohr P, Schroll M, Hein HO. All-cause mortality associated with physical activity during leisure time, work, sports, and cycling to work. Arch Intern Med 160: 1621–1628, 2000. doi:10.1001/archinte.160.11.1621.
    Crossref | PubMed | Web of Science | Google Scholar
  • 857. Moore SC, Lee IM, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM, , et al.. Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med 176: 816–825, 2016. doi:10.1001/jamainternmed.2016.1548.
    Crossref | PubMed | Web of Science | Google Scholar
  • 858. Lavery JA, Boutros PC, Knight D, Tammela T, Moskowitz CS, Jones LW. Association of exercise with pan-cancer incidence and overall survival. Cancer Cell 42: 169–171, 2024. doi:10.1016/j.ccell.2023.12.007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 859. Matthews CE, Moore SC, Arem H, Cook MB, Trabert B, Håkansson N, Larsson SC, Wolk A, Gapstur SM, Lynch BM, Milne RL, Freedman ND, Huang WY, Berrington de Gonzalez A, Kitahara CM, Linet MS, Shiroma EJ, Sandin S, Patel AV, Lee IM. Amount and intensity of leisure-time physical activity and lower cancer risk. J Clin Oncol 38: 686–697, 2020. doi:10.1200/JCO.19.02407.
    Crossref | PubMed | Web of Science | Google Scholar
  • 860. Liu L, Shi Y, Li T, Qin Q, Yin J, Pang S, Nie S, Wei S. Leisure time physical activity and cancer risk: evaluation of the WHO’s recommendation based on 126 high-quality epidemiological studies. Br J Sports Med 50: 372–378, 2016. doi:10.1136/bjsports-2015-094728.
    Crossref | PubMed | Web of Science | Google Scholar
  • 861. Fiuza-Luces C, Valenzuela PL, Gálvez BG, Ramírez M, López-Soto A, Simpson RJ, Lucia A. The effect of physical exercise on anticancer immunity. Nat Rev Immunol 24: 282–293, 2024. doi:10.1038/s41577-023-00943-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 862. Dighriri A, Timraz M, Rosaini ND, Aba Alkhayl FF, Boyle JG, Logan G, Gray SR. The impact of the time of day on metabolic responses to exercise in adults: a systematic and meta-analysis review. Chronobiol Int 41: 1377–1388, 2024. doi:10.1080/07420528.2024.2419867.
    Crossref | PubMed | Web of Science | Google Scholar
  • 863. Feng H, Yang L, Liang YY, Ai S, Liu Y, Liu Y, Jin X, Lei B, Wang J, Zheng N, Chen X, Chan JW, Sum RK, Chan NY, Tan X, Benedict C, Wing YK, Zhang J. Associations of timing of physical activity with all-cause and cause-specific mortality in a prospective cohort study. Nat Commun 14: 930, 2023. doi:10.1038/s41467-023-36546-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 864. Polo-López A, Calatayud J, Palau P, López-Bueno L, Nuñez-Cortés R, Andersen LL, López-Bueno R. Joint associations of handgrip strength and physical activity with incident cardiovascular disease and overall mortality in the UK Biobank. Clin Nutr 43: 218–224, 2024. doi:10.1016/j.clnu.2024.10.022.
    Crossref | PubMed | Web of Science | Google Scholar
  • 865. Dunstan DW, Dogra S, Carter SE, Owen N. Sit less and move more for cardiovascular health: emerging insights and opportunities. Nat Rev Cardiol 18: 637–648, 2021. doi:10.1038/s41569-021-00547-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 866. Pinto AJ, Bergouignan A, Dempsey PC, Roschel H, Owen N, Gualano B, Dunstan DW. Physiology of sedentary behavior. Physiol Rev 103: 2561–2622, 2023. doi:10.1152/physrev.00022.2022.
    Link | Web of Science | Google Scholar
  • 867. Biswas A, Oh PI, Faulkner GE, Bajaj RR, Silver MA, Mitchell MS, Alter DA. Sedentary time and its association with risk for disease incidence, mortality, and hospitalization in adults: a systematic review and meta-analysis. Ann Intern Med 162: 123–132, 2015. doi:10.7326/M14-1651.
    Crossref | PubMed | Web of Science | Google Scholar
  • 868. Guo C, Zhou Q, Zhang D, Qin P, Li Q, Tian G, Liu D, Chen X, Liu L, Liu F, Cheng C, Qie R, Han M, Huang S, Wu X, Zhao Y, Ren Y, Zhang M, Liu Y, Hu D. Association of total sedentary behaviour and television viewing with risk of overweight/obesity, type 2 diabetes and hypertension: a dose-response meta-analysis. Diabetes Obes Metab 22: 79–90, 2020. doi:10.1111/dom.13867.
    Crossref | PubMed | Web of Science | Google Scholar
  • 869. Wilmot EG, Edwardson CL, Achana FA, Davies MJ, Gorely T, Gray LJ, Khunti K, Yates T, Biddle SJ. Sedentary time in adults and the association with diabetes, cardiovascular disease and death: systematic review and meta-analysis. Diabetologia 55: 2895–2905, 2012. doi:10.1007/s00125-012-2677-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 870. Ekelund U, Tarp J, Fagerland MW, Johannessen JS, Hansen BH, Jefferis BJ, Whincup PH, Diaz KM, Hooker S, Howard VJ, Chernofsky A, Larson MG, Spartano N, Vasan RS, Dohrn IM, Hagströmer M, Edwardson C, Yates T, Shiroma EJ, Dempsey P, Wijndaele K, Anderssen SA, Lee IM. Joint associations of accelero-meter measured physical activity and sedentary time with all-cause mortality: a harmonised meta-analysis in more than 44 000 middle-aged and older individuals. Br J Sports Med 54: 1499–1506, 2020. doi:10.1136/bjsports-2020-103270.
    Crossref | PubMed | Web of Science | Google Scholar
  • 871. Henson J, De Craemer M, Yates T. Sedentary behaviour and disease risk. BMC Public Health 23: 2048, 2023. doi:10.1186/s12889-023-16867-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 872. Matthews CE, George SM, Moore SC, Bowles HR, Blair A, Park Y, Troiano RP, Hollenbeck A, Schatzkin A. Amount of time spent in sedentary behaviors and cause-specific mortality in US adults. Am J Clin Nutr 95: 437–445, 2012. doi:10.3945/ajcn.111.019620.
    Crossref | PubMed | Web of Science | Google Scholar
  • 873. Li S, Lear SA, Rangarajan S, Hu B, Yin L, Bangdiwala SI, , et al.. Association of sitting time with mortality and cardiovascular events in high-income, middle-income, and low-income countries. JAMA Cardiol 7: 796–807, 2022. doi:10.1001/jamacardio.2022.1581.
    Crossref | PubMed | Web of Science | Google Scholar
  • 874. Patterson R, McNamara E, Tainio M, de Sá TH, Smith AD, Sharp SJ, Edwards P, Woodcock J, Brage S, Wijndaele K. Sedentary behaviour and risk of all-cause, cardiovascular and cancer mortality, and incident type 2 diabetes: a systematic review and dose response meta-analysis. Eur J Epidemiol 33: 811–829, 2018. doi:10.1007/s10654-018-0380-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 875. Ekelund U, Brown WJ, Steene-Johannessen J, Fagerland MW, Owen N, Powell KE, Bauman AE, Lee IM. Do the associations of sedentary behaviour with cardiovascular disease mortality and cancer mortality differ by physical activity level? A systematic review and harmonised meta-analysis of data from 850 060 participants. Br J Sports Med 53: 886–894, 2019. doi:10.1136/bjsports-2017-098963.
    Crossref | PubMed | Web of Science | Google Scholar
  • 876. Zhao R, Bu W, Chen Y, Chen X. The dose-response associations of sedentary time with chronic diseases and the risk for all-cause mortality affected by different health status: a systematic review and meta-analysis. J Nutr Health Aging 24: 63–70, 2020. doi:10.1007/s12603-019-1298-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 877. Ekelund U, Steene-Johannessen J, Brown WJ, Fagerland MW, Owen N, Powell KE, Bauman A, Lee IM; Lancet Physical Activity Series 2 Executive Committee, Lancet Sedentary Behaviour Working Group. Does physical activity attenuate, or even eliminate, the detrimental association of sitting time with mortality? A harmonised meta-analysis of data from more than 1 million men and women. Lancet 388: 1302–1310, 2016. doi:10.1016/S0140-6736(16)30370-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 878. Ahmadi MN, Blodgett JM, Atkin AJ, Chan HW, Del Pozo Cruz B, Suorsa K, Bakker EA, Pulsford RM, Mielke GI, Johansson PJ, Hettiarachchi P, Thijssen DH, Stenholm S, Mishra GD, Teixeira-Pinot A, Rangul V, Sherar LB, Ekelund U, Hughes AD, Lee IM, Holtermann A, Koster A, Hamer M, Stamatakis E; ProPASS collaboration. Relationship of device measured physical activity type and posture with cardiometabolic health markers: pooled dose-response associations from the Prospective Physical Activity, Sitting and Sleep Consortium. Diabetologia 67: 1051–1065, 2024. doi:10.1007/s00125-024-06090-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 879. Koemel NA, Ahmadi MN, Biswas RK, Koster A, Atkin AJ, Sabag A, Stamatakis E. Can incidental physical activity offset the deleterious associations of sedentary behaviour with major adverse cardiovascular events? Eur J Prev Cardiol 32: 77–85, 2025. doi:10.1093/eurjpc/zwae316.
    Crossref | PubMed | Web of Science | Google Scholar
  • 880. Rezende LF, Ahmadi M, Ferrari G, Del Pozo Cruz B, Lee IM, Ekelund U, Stamatakis E. Device-measured sedentary time and intensity-specific physical activity in relation to all-cause and cardiovascular disease mortality: the UK Biobank cohort study. Int J Behav Nutr Phys Act 21: 68, 2024. doi:10.1186/s12966-024-01615-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 881. Brakenridge CJ, Koster A, de Galan BE, Carver A, Dumuid D, Dzakpasu FQ, Eussen S, Savelberg H, Bosma H, Owen N, Schaper NC, Healy GN, Dunstan DW. Associations of 24 h time-use compositions of sitting, standing, physical activity and sleeping with optimal cardiometabolic risk and glycaemic control: the Maastricht Study. Diabetologia 67: 1356–1367, 2024. doi:10.1007/s00125-024-06145-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 882. Smirnova E, Leroux A, Cao Q, Tabacu L, Zipunnikov V, Crainiceanu C, Urbanek JK. The predictive performance of objective measures of physical activity derived from accelerometry data for 5-year all-cause mortality in older adults: National Health and Nutritional Examination Survey 2003-2006. J Gerontol A Biol Sci Med Sci 75: 1779–1785, 2020. doi:10.1093/gerona/glz193.
    Crossref | PubMed | Web of Science | Google Scholar
  • 883. Pyrkov TV, Getmantsev E, Zhurov B, Avchaciov K, Pyatnitskiy M, Menshikov L, Khodova K, Gudkov AV, Fedichev PO. Quantitative characterization of biological age and frailty based on locomotor activity records. Aging (Albany NY) 10: 2973–2990, 2018. doi:10.18632/aging.101603.
    Crossref | PubMed | Web of Science | Google Scholar
  • 884. Ekelund U, Tarp J, Sanchez-Lastra MA, Dalene KE. Physical activity, sedentary time and health - a narrative review with new insights. Dan Med J 71: A06240433, 2024. doi:10.61409/A06240433.
    Crossref | PubMed | Web of Science | Google Scholar
  • 885. Banach M, Lewek J, Surma S, Penson PE, Sahebkar A, Martin SS, Bajraktari G, Henein MY, Reiner Z, Bielecka-Dabrowa A, Bytyçi I. The association between daily step count and all-cause and cardiovascular mortality: a meta-analysis. Eur J Prev Cardiol 30: 1975–1985, 2023. doi:10.1093/eurjpc/zwad229.
    Crossref | PubMed | Web of Science | Google Scholar
  • 886. Lee IM, Shiroma EJ, Kamada M, Bassett DR, Matthews CE, Buring JE. Association of step volume and intensity with all-cause mortality in older women. JAMA Intern Med 179: 1105–1112, 2019. doi:10.1001/jamainternmed.2019.0899.
    Crossref | PubMed | Web of Science | Google Scholar
  • 887. Master H, Annis J, Huang S, Beckman JA, Ratsimbazafy F, Marginean K, Carroll R, Natarajan K, Harrell FE, Roden DM, Harris P, Brittain EL. Association of step counts over time with the risk of chronic disease in the All of Us Research Program. Nat Med 28: 2301–2308, 2022. doi:10.1038/s41591-022-02012-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 888. Paluch AE, Bajpai S, Bassett DR, Carnethon MR, Ekelund U, Evenson KR, Steps for Health C, , et al.. Daily steps and all-cause mortality: a meta-analysis of 15 international cohorts. Lancet Public Health 7: e219–e228, 2022. doi:10.1016/S2468-2667(21)00302-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 889. Ramsey KA, Meskers CG, Maier AB. Every step counts: synthesising reviews associating objectively measured physical activity and sedentary behaviour with clinical outcomes in community-dwelling older adults. Lancet Healthy Longev 2: e764–e772, 2021. doi:10.1016/S2666-7568(21)00203-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 890. Saint-Maurice PF, Troiano RP, Bassett DR Jr, Graubard BI, Carlson SA, Shiroma EJ, Fulton JE, Matthews CE. Association of daily step count and step intensity with mortality among US adults. JAMA 323: 1151–1160, 2020. doi:10.1001/jama.2020.1382.
    Crossref | PubMed | Web of Science | Google Scholar
  • 891. Stens NA, Bakker EA, Mañas A, Buffart LM, Ortega FB, Lee DC, Thompson PD, Thijssen DH, Eijsvogels TM. Relationship of daily step counts to all-cause mortality and cardiovascular events. J Am Coll Cardiol 82: 1483–1494, 2023. doi:10.1016/j.jacc.2023.07.029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 892. Ahmadi MN, Rezende LF, Ferrari G, Del Pozo Cruz B, Lee IM, Stamatakis E. Do the associations of daily steps with mortality and incident cardiovascular disease differ by sedentary time levels? A device-based cohort study. Br J Sports Med 58: 261–268, 2024. doi:10.1136/bjsports-2023-107221.
    Crossref | PubMed | Web of Science | Google Scholar
  • 893. Xu C, Jia J, Zhao B, Yuan M, Luo N, Zhang F, Wang H. Objectively measured daily steps and health outcomes: an umbrella review of the systematic review and meta-analysis of observational studies. BMJ Open 14: e088524, 2024. doi:10.1136/bmjopen-2024-088524.
    Crossref | PubMed | Web of Science | Google Scholar
  • 894. Del Pozo Cruz B, Ahmadi MN, Lee IM, Stamatakis E. Prospective associations of daily step counts and intensity with cancer and cardiovascular disease incidence and mortality and all-cause mortality. JAMA Intern Med 182: 1139–1148, 2022. doi:10.1001/jamainternmed.2022.4000.
    Crossref | PubMed | Web of Science | Google Scholar
  • 895. Del Pozo Cruz B, Ahmadi M, Naismith SL, Stamatakis E. Association of daily step count and intensity with incident dementia in 78 430 adults living in the UK. JAMA Neurol 79: 1059–1063, 2022. doi:10.1001/jamaneurol.2022.2672.
    Crossref | PubMed | Web of Science | Google Scholar
  • 896. Bizzozero-Peroni B, Diaz-Goni V, Jimenez-Lopez E, Rodriguez-Gutierrez E, Sequi-Dominguez I, Nunez de Arenas-Arroyo S, Lopez-Gil JF, Martinez-Vizcaino V, Mesas AE. Daily step count and depression in adults: a systematic review and meta-analysis. JAMA Netw Open 7: e2451208, 2024. doi:10.1001/jamanetworkopen.2024.51208.
    Crossref | PubMed | Web of Science | Google Scholar
  • 897. Stamatakis E, Ahmadi MN, Friedenreich CM, Blodgett JM, Koster A, Holtermann A, Atkin A, Rangul V, Sherar LB, Teixeira-Pinto A, Ekelund U, Lee IM, Hamer M. Vigorous intermittent lifestyle physical activity and cancer incidence among nonexercising adults: the UK Biobank Accelerometry Study. JAMA Oncol 9: 1255–1259, 2023. doi:10.1001/jamaoncol.2023.1830.
    Crossref | PubMed | Web of Science | Google Scholar
  • 898. Stamatakis E, Ahmadi MN, Gill JM, Thøgersen-Ntoumani C, Gibala MJ, Doherty A, Hamer M. Association of wearable device-measured vigorous intermittent lifestyle physical activity with mortality. Nat Med 28: 2521–2529, 2022. doi:10.1038/s41591-022-02100-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 899. Ekelund U, Tarp J, Steene-Johannessen J, Hansen BH, Jefferis B, Fagerland MW, Whincup P, Diaz KM, Hooker SP, Chernofsky A, Larson MG, Spartano N, Vasan RS, Dohrn IM, Hagstromer M, Edwardson C, Yates T, Shiroma E, Anderssen SA, Lee IM. Dose-response associations between accelerometry measured physical activity and sedentary time and all cause mortality: systematic review and harmonised meta-analysis. BMJ 366: l4570, 2019. doi:10.1136/bmj.l4570.
    Crossref | PubMed | Web of Science | Google Scholar
  • 900. Nes BM, Gutvik CR, Lavie CJ, Nauman J, Wisløff U. Personalized activity intelligence (PAI) for prevention of cardiovascular disease and promotion of physical activity. Am J Med 130: 328–336, 2017. doi:10.1016/j.amjmed.2016.09.031.
    Crossref | PubMed | Web of Science | Google Scholar
  • 901. Nauman J, Franklin BA, Nes BM, Sallis RE, Sawada SS, Marinović J, Stensvold D, Lavie CJ, Tari AR, Wisløff U. Association between personal activity intelligence and mortality: population-based China Kadoorie Biobank Study. Mayo Clin Proc 97: 668–681, 2022. doi:10.1016/j.mayocp.2021.10.022.
    Crossref | PubMed | Web of Science | Google Scholar
  • 902. Stamatakis E, Ekelund U, Ding D, Hamer M, Bauman AE, Lee IM. Is the time right for quantitative public health guidelines on sitting? A narrative review of sedentary behaviour research paradigms and findings. Br J Sports Med 53: 377–382, 2019. doi:10.1136/bjsports-2018-099131.
    Crossref | PubMed | Web of Science | Google Scholar
  • 903. Doherty C, Baldwin M, Keogh A, Caulfield B, Argent R. Keeping pace with wearables: a living umbrella review of systematic reviews evaluating the accuracy of consumer wearable technologies in health measurement. Sports Med 54: 2907–2926, 2024. doi:10.1007/s40279-024-02077-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 904. Shailendra P, Baldock KL, Li LS, Bennie JA, Boyle T. Resistance training and mortality risk: a systematic review and meta-analysis. Am J Prev Med 63: 277–285, 2022. doi:10.1016/j.amepre.2022.03.020.
    Crossref | PubMed | Web of Science | Google Scholar
  • 905. Westcott WL. Resistance training is medicine: effects of strength training on health. Curr Sports Med Rep 11: 209–216, 2012. doi:10.1249/JSR.0b013e31825dabb8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 906. Fragala MS, Cadore EL, Dorgo S, Izquierdo M, Kraemer WJ, Peterson MD, Ryan ED. Resistance training for older adults: position statement from the National Strength and Conditioning Association. J Strength Cond Res 33: 2019–2052, 2019. doi:10.1519/JSC.0000000000003230.
    Crossref | PubMed | Web of Science | Google Scholar
  • 907. Liu Y, Lee DC, Li Y, Zhu W, Zhang R, Sui X, Lavie CJ, Blair SN. Associations of resistance exercise with cardiovascular disease morbidity and mortality. Med Sci Sports Exerc 51: 499–508, 2019. doi:10.1249/MSS.0000000000001822.
    Crossref | PubMed | Web of Science | Google Scholar
  • 908. Momma H, Kawakami R, Honda T, Sawada SS. Muscle-strengthening activities are associated with lower risk and mortality in major non-communicable diseases: a systematic review and meta-analysis of cohort studies. Br J Sports Med 56: 755–763, 2022. doi:10.1136/bjsports-2021-105061.
    Crossref | PubMed | Web of Science | Google Scholar
  • 909. Kang DW, Dawson JK, Barnes O, Wilson RL, Norris MK, Gonzalo-Encabo P, Christopher CN, Ficarra S, Dieli-Conwright CM. Resistance exercise and skeletal muscle-related outcomes in patients with cancer: a systematic review. Med Sci Sports Exerc 56: 1747–1758, 2024. doi:10.1249/MSS.0000000000003452.
    Crossref | PubMed | Web of Science | Google Scholar
  • 910. Abou Sawan S, Nunes EA, Lim C, McKendry J, Phillips SM. The health benefits of resistance exercise: beyond hypertrophy and big weights. Exerc Sport Mov 1: e00001, 2023. doi:10.1249/ESM.0000000000000001.
    Crossref | Google Scholar
  • 911. Choi Y, Kim D, Kim SK. Effects of physical activity on body composition, muscle strength, and physical function in old age: bibliometric and meta-analyses. Healthcare (Basel) 12, 2024. doi:10.3390/healthcare12020197.
    Crossref | PubMed | Web of Science | Google Scholar
  • 912. Saeidifard F, Medina-Inojosa JR, West CP, Olson TP, Somers VK, Bonikowske AR, Prokop LJ, Vinciguerra M, Lopez-Jimenez F. The association of resistance training with mortality: a systematic review and meta-analysis. Eur J Prev Cardiol 26: 1647–1665, 2019. doi:10.1177/2047487319850718.
    Crossref | PubMed | Web of Science | Google Scholar
  • 913. Stamatakis E, Lee IM, Bennie J, Freeston J, Hamer M, O’Donovan G, Ding D, Bauman A, Mavros Y. Does strength-promoting exercise confer unique health benefits? A pooled analysis of data on 11 population cohorts with all-cause, cancer, and cardiovascular mortality endpoints. Am J Epidemiol 187: 1102–1112, 2018. doi:10.1093/aje/kwx345.
    Crossref | PubMed | Web of Science | Google Scholar
  • 914. Bennie JA, Shakespear-Druery J, De Cocker K. Muscle-strengthening exercise epidemiology: a new frontier in chronic disease prevention. Sports Med Open 6: 40, 2020. doi:10.1186/s40798-020-00271-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 915. Paluch AE, Boyer WR, Franklin BA, Laddu D, Lobelo F, Lee DC, McDermott MM, Swift DL, Webel AR, Lane A; American Heart Association Council on Lifestyle and Cardiometabolic Health, Council on Arteriosclerosis, Thrombosis and Vascular Biology, Council on Clinical Cardiology, Council on Cardiovascular and Stroke Nursing, Council on Epidemiology and Prevention, Council on Peripheral Vascular Disease. Resistance Exercise Training in Individuals With and Without Cardiovascular Disease: 2023 Update: a scientific statement from the American Heart Association. Circulation 149: e217–e231, 2024. doi:10.1161/CIR.0000000000001189.
    Crossref | PubMed | Web of Science | Google Scholar
  • 916. Hardee JP, Porter RR, Sui X, Archer E, Lee IM, Lavie CJ, Blair SN. The effect of resistance exercise on all-cause mortality in cancer survivors. Mayo Clin Proc 89: 1108–1115, 2014. doi:10.1016/j.mayocp.2014.03.018.
    Crossref | PubMed | Web of Science | Google Scholar
  • 917. Kraschnewski JL, Sciamanna CN, Poger JM, Rovniak LS, Lehman EB, Cooper AB, Ballentine NH, Ciccolo JT. Is strength training associated with mortality benefits? A 15year cohort study of US older adults. Prev Med 87: 121–127, 2016. doi:10.1016/j.ypmed.2016.02.038.
    Crossref | PubMed | Web of Science | Google Scholar
  • 918. Cunha PM, Werneck AO, Santos LD, Oliveira MD, Zou L, Schuch FB, Cyrino ES. Can resistance training improve mental health outcomes in older adults? A systematic review and meta-analysis of randomized controlled trials. Psychiatry Res 333: 115746, 2024. doi:10.1016/j.psychres.2024.115746.
    Crossref | PubMed | Web of Science | Google Scholar
  • 919. Giovannucci EL, Rezende LF, Lee DH. Muscle-strengthening activities and risk of cardiovascular disease, type 2 diabetes, cancer and mortality: a review of prospective cohort studies. J Intern Med 290: 789–805, 2021. doi:10.1111/joim.13344.
    Crossref | PubMed | Web of Science | Google Scholar
  • 920. Bennie JA, Lee DC, Brellenthin AG, De Cocker K. Muscle-strengthening exercise and prevalent hypertension among 1.5 million adults: a little is better than none. J Hypertens 38: 1466–1473, 2020. doi:10.1097/HJH.0000000000002415.
    Crossref | PubMed | Web of Science | Google Scholar
  • 921. Currier BS, McLeod JC, Banfield L, Beyene J, Welton NJ, D’Souza AC, Keogh JA, Lin L, Coletta G, Yang A, Colenso-Semple L, Lau KJ, Verboom A, Phillips SM. Resistance training prescription for muscle strength and hypertrophy in healthy adults: a systematic review and Bayesian network meta-analysis. Br J Sports Med 57: 1211–1220, 2023. doi:10.1136/bjsports-2023-106807.
    Crossref | PubMed | Web of Science | Google Scholar
  • 922. Olsen RJ, Hasan SS, Woo JJ, Nawabi DH, Ramkumar PN. The fundamentals and applications of wearable sensor devices in sports medicine: a scoping review. Arthroscopy 41: 473–492, 2025. doi:10.1016/j.arthro.2024.01.042.
    Crossref | PubMed | Web of Science | Google Scholar
  • 923. Kim Y, White T, Wijndaele K, Westgate K, Sharp SJ, Helge JW, Wareham NJ, Brage S. The combination of cardiorespiratory fitness and muscle strength, and mortality risk. Eur J Epidemiol 33: 953–964, 2018. doi:10.1007/s10654-018-0384-x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 924. Brellenthin AG, Bennie JA, Lee DC. Aerobic or muscle-strengthening physical activity: which is better for health? Curr Sports Med Rep 21: 272–279, 2022. doi:10.1249/JSR.0000000000000981.
    Crossref | PubMed | Web of Science | Google Scholar
  • 925. O’Keefe JH, O’Keefe EL, Eckert R, Lavie CJ. Training strategies to optimize cardiovascular durability and life expectancy. Mo Med 120: 155–162, 2023. https://pmc.ncbi.nlm.nih.gov/articles/PMC10121111/.
    PubMed | Google Scholar
  • 926. Coleman CJ, McDonough DJ, Pope ZC, Pope CA. Dose-response association of aerobic and muscle-strengthening physical activity with mortality: a national cohort study of 416 420 US adults. Br J Sports Med 2022: bjsports-2022-105519, 2022. doi:10.1136/bjsports-2022-105519.
    Crossref | PubMed | Web of Science | Google Scholar
  • 927. Marzola P, Melzer T, Pavesi E, Gil-Mohapel J, Brocardo PS. Exploring the role of neuroplasticity in development, aging, and neurodegeneration. Brain Sci 13: 1610, 2023. doi:10.3390/brainsci13121610.
    Crossref | PubMed | Web of Science | Google Scholar
  • 928. Lee J, Kim HJ. Normal aging induces changes in the brain and neurodegeneration progress: review of the structural, biochemical, metabolic, cellular, and molecular changes. Front Aging Neurosci 14: 931536, 2022. doi:10.3389/fnagi.2022.931536.
    Crossref | PubMed | Web of Science | Google Scholar
  • 929. Tsai SY. Lost in translation: challenges of current pharmacotherapy for sarcopenia. Trends Mol Med 30: 1047–1060, 2024. doi:10.1016/j.molmed.2024.05.016.
    Crossref | PubMed | Web of Science | Google Scholar
  • 930. Boccardi V. Sarcopenia: A dive into metabolism to promote a multimodal, preventive, and regenerative approach. Mech Ageing Dev 219: 111941, 2024. doi:10.1016/j.mad.2024.111941.
    Crossref | PubMed | Web of Science | Google Scholar
  • 931. Larsson L, Degens H, Li M, Salviati L, Lee YI, Thompson W, Kirkland JL, Sandri M. Sarcopenia: aging-related loss of muscle mass and function. Physiol Rev 99: 427–511, 2019. doi:10.1152/physrev.00061.2017.
    Link | Web of Science | Google Scholar
  • 932. Sayer AA, Cooper R, Arai H, Cawthon PM, Ntsama Essomba MJ, Fielding RA, Grounds MD, Witham MD, Cruz-Jentoft AJ. Sarcopenia. Nat Rev Dis Primers 10: 68, 2024. doi:10.1038/s41572-024-00550-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 933. Miri S, Farhadi B, Takasi P, Ghorbani Vajargah P, Karkhah S. Physical independence and related factors among older adults: a systematic review and meta-analysis. Ann Med Surg (Lond) 86: 3400–3408, 2024. doi:10.1097/MS9.0000000000002100.
    Crossref | PubMed | Web of Science | Google Scholar
  • 934. Fleg JL, Strait J. Age-associated changes in cardiovascular structure and function: a fertile milieu for future disease. Heart Fail Rev 17: 545–554, 2012. doi:10.1007/s10741-011-9270-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 935. Strait JB, Lakatta EG. Aging-associated cardiovascular changes and their relationship to heart failure. Heart Fail Clin 8: 143–164, 2012. doi:10.1016/j.hfc.2011.08.011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 936. Murray KO, Mahoney SA, Venkatasubramanian R, Seals DR, Clayton ZS. Aging, aerobic exercise, and cardiovascular health: barriers, alternative strategies and future directions. Exp Gerontol 173: 112105, 2023. doi:10.1016/j.exger.2023.112105.
    Crossref | PubMed | Web of Science | Google Scholar
  • 937. Ahmed B, Rahman AA, Lee S, Malhotra R. The implications of aging on vascular health. Int J Mol Sci 25: 11188, 2024. doi:10.3390/ijms252011188.
    Crossref | PubMed | Web of Science | Google Scholar
  • 938. Boskey AL, Imbert L. Bone quality changes associated with aging and disease: a review. Ann NY Acad Sci 1410: 93–106, 2017. doi:10.1111/nyas.13572.
    Crossref | PubMed | Web of Science | Google Scholar
  • 939. Mohebbi R, Shojaa M, Kohl M, von Stengel S, Jakob F, Kerschan-Schindl K, Lange U, Peters S, Thomasius F, Uder M, Kemmler W. Exercise training and bone mineral density in postmenopausal women: an updated systematic review and meta-analysis of intervention studies with emphasis on potential moderators. Osteoporos Int 34: 1145–1178, 2023. doi:10.1007/s00198-023-06682-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 940. David K, Narinx N, Antonio L, Evenepoel P, Claessens F, Decallonne B, Vanderschueren D. Bone health in ageing men. Rev Endocr Metab Disord 23: 1173–1208, 2022. doi:10.1007/s11154-022-09738-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 941. Cheng N, Josse AR. Dairy and exercise for bone health: evidence from randomized controlled trials and recommendations for future research. Curr Osteoporos Rep 22: 502–514, 2024. doi:10.1007/s11914-024-00882-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 942. Ning Y, Chen M, An J, Tang M, Tse G, Chan JSK, Zhao C, Liu Y, Lei X, Qiang H, Bai C, Li H, Yu H, Yan Y, Wang D, Li G. Association between “weekend warrior” physical activity and the incidence of neurodegenerative diseases. Neurotherapeutics 21: e00430, 2024. doi:10.1016/j.neurot.2024.e00430.
    Crossref | PubMed | Web of Science | Google Scholar
  • 943. Sánchez-Sánchez JL, He L, Morales JS, de Souto Barreto P, Jiménez-Pavón D, Carbonell-Baeza A, Casas-Herrero A, Gallardo-Gómez D, Lucia A, Del Pozo Cruz B, Valenzuela PL. Association of physical behaviours with sarcopenia in older adults: a systematic review and meta-analysis of observational studies. Lancet Healthy Longev 5: e108–e119, 2024. doi:10.1016/S2666-7568(23)00241-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 944. McKendry J, Coletta G, Nunes EA, Lim C, Phillips SM. Mitigating disuse-induced skeletal muscle atrophy in ageing: resistance exercise as a critical countermeasure. Exp Physiol 109: 1650–1662, 2024. doi:10.1113/EP091937.
    Crossref | PubMed | Web of Science | Google Scholar
  • 945. Ding L, Yang F. Muscle weakness is related to slip-initiated falls among community-dwelling older adults. J Biomech 49: 238–243, 2016. doi:10.1016/j.jbiomech.2015.12.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 946. Gauvain JB, Mandigout S, Pambet M, Monseu M, Gillain P, Gautier J, Annweiler C, Puisieux F. Correlation between muscle mass and physical activity level in older adults at risk of falling: the FITNESS Study. J Frailty Aging 13: 240–247, 2024. doi:10.14283/jfa.2024.53.
    Crossref | PubMed | Web of Science | Google Scholar
  • 947. Rodrigues F, Domingos C, Monteiro D, Morouço P. A review on aging, sarcopenia, falls, and resistance training in community-dwelling older adults. Int J Environ Res Public Health 19: 874, 2022. doi:10.3390/ijerph19020874.
    Crossref | PubMed | Web of Science | Google Scholar
  • 948. English KL, Paddon-Jones D. Protecting muscle mass and function in older adults during bed rest. Curr Opin Clin Nutr Metab Care 13: 34–39, 2010. doi:10.1097/MCO.0b013e328333aa66.
    Crossref | PubMed | Web of Science | Google Scholar
  • 949. Aldrich L, Ispoglou T, Prokopidis K, Alqallaf J, Wilson O, Stavropoulos-Kalinoglou A. Acute sarcopenia: systematic review and meta-analysis on its incidence and muscle parameter shifts during hospitalisation. J Cachexia Sarcopenia Muscle 16: e13662, 2025. doi:10.1002/jcsm.13662.
    Crossref | PubMed | Web of Science | Google Scholar
  • 950. Welch C, Chen Y, Hartley P, Naughton C, Martinez-Velilla N, Stein D, Romero-Ortuno R. New horizons in hospital-associated deconditioning: a global condition of body and mind. Age Ageing 53: afae241, 2024. doi:10.1093/ageing/afae241.
    Crossref | PubMed | Web of Science | Google Scholar
  • 951. Liu J, Xue H, Ma YH, Wang Z. Acute muscle loss in elderly hospitalized patients: Risk factors and adverse clinical outcomes. Geriatr Nurs 61: 449–454, 2025. doi:10.1016/j.gerinurse.2024.12.032.
    Crossref | PubMed | Web of Science | Google Scholar
  • 952. Zanker J, Scott D, Alajlouni D, Kirk B, Bird S, DeBruin D, Vogrin S, Bliuc D, Tran T, Cawthon P, Duque G, Center JR. Mortality, falls and slow walking speed are predicted by different muscle strength and physical performance measures in women and men. Arch Gerontol Geriatr 114: 105084, 2023. doi:10.1016/j.archger.2023.105084.
    Crossref | PubMed | Web of Science | Google Scholar
  • 953. White DK, Neogi T, Nevitt MC, Peloquin CE, Zhu Y, Boudreau RM, Cauley JA, Ferrucci L, Harris TB, Satterfield SM, Simonsick EM, Strotmeyer ES, Zhang Y. Trajectories of gait speed predict mortality in well-functioning older adults: the Health, Aging and Body Composition study. J Gerontol A Biol Sci Med Sci 68: 456–464, 2013. doi:10.1093/gerona/gls197.
    Crossref | PubMed | Web of Science | Google Scholar
  • 954. Shinkai S, Watanabe S, Kumagai S, Fujiwara Y, Amano H, Yoshida H, Ishizaki T, Yukawa H, Suzuki T, Shibata H. Walking speed as a good predictor for the onset of functional dependence in a Japanese rural community population. Age Ageing 29: 441–446, 2000. doi:10.1093/ageing/29.5.441.
    Crossref | PubMed | Web of Science | Google Scholar
  • 955. Howlett SE, Rutenberg AD, Rockwood K. The degree of frailty as a translational measure of health in aging. Nat Aging 1: 651–665, 2021. doi:10.1038/s43587-021-00099-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 956. Studenski S, Perera S, Patel K, Rosano C, Faulkner K, Inzitari M, Brach J, Chandler J, Cawthon P, Connor EB, Nevitt M, Visser M, Kritchevsky S, Badinelli S, Harris T, Newman AB, Cauley J, Ferrucci L, Guralnik J. Gait speed and survival in older adults. JAMA 305: 50–58, 2011. doi:10.1001/jama.2010.1923.
    Crossref | PubMed | Web of Science | Google Scholar
  • 957. Stanaway FF, Gnjidic D, Blyth FM, Le Couteur DG, Naganathan V, Waite L, Seibel MJ, Handelsman DJ, Sambrook PN, Cumming RG. How fast does the Grim Reaper walk? Receiver operating characteristics curve analysis in healthy men aged 70 and over. BMJ 343: d7679, 2011. doi:10.1136/bmj.d7679.
    Crossref | PubMed | Web of Science | Google Scholar
  • 958. Lo AX, Donnelly JP, McGwin G Jr, Bittner V, Ahmed A, Brown CJ. Impact of gait speed and instrumental activities of daily living on all-cause mortality in adults >/=65 years with heart failure. Am J Cardiol 115: 797–801, 2015. doi:10.1016/j.amjcard.2014.12.044.
    Crossref | PubMed | Web of Science | Google Scholar
  • 959. Sonn U. Longitudinal studies of dependence in daily life activities among elderly persons. Scand J Rehabil Med Suppl 34: 1–35, 1996.
    PubMed | Web of Science | Google Scholar
  • 960. Elbaz A, Sabia S, Brunner E, Shipley M, Marmot M, Kivimaki M, Singh-Manoux A. Association of walking speed in late midlife with mortality: results from the Whitehall II cohort study. Age (Dordr) 35: 943–952, 2013. doi:10.1007/s11357-012-9387-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 961. Franklin BA, Brinks J, Sacks R, Trivax J, Friedman H. Reduced walking speed and distance as harbingers of the approaching grim reaper. Am J Cardiol 116: 313–317, 2015. doi:10.1016/j.amjcard.2015.04.024.
    Crossref | PubMed | Web of Science | Google Scholar
  • 962. Boyer KA, Hayes KL, Umberger BR, Adamczyk PG, Bean JF, Brach JS, Clark BC, Clark DJ, Ferrucci L, Finley J, Franz JR, Golightly YM, Hortobágyi T, Hunter S, Narici M, Nicklas B, Roberts T, Sawicki G, Simonsick E, Kent JA. Age-related changes in gait biomechanics and their impact on the metabolic cost of walking: report from a National Institute on Aging workshop. Exp Gerontol 173: 112102, 2023. doi:10.1016/j.exger.2023.112102.
    Crossref | PubMed | Web of Science | Google Scholar
  • 963. Gamwell HE, Wait SO, Royster JT, Ritch BL, Powell SC, Skinner JW. Aging and gait function: examination of multiple factors that influence gait variability. Gerontol Geriatr Med 8: 23337214221080304, 2022. doi:10.1177/23337214221080304.
    Crossref | PubMed | Web of Science | Google Scholar
  • 964. Hulleck AA, Menoth Mohan D, Abdallah N, El Rich M, Khalaf K. Present and future of gait assessment in clinical practice: towards the application of novel trends and technologies. Front Med Technol 4: 901331, 2022. doi:10.3389/fmedt.2022.901331.
    Crossref | PubMed | Web of Science | Google Scholar
  • 965. Seo S, Kwon T, Shin M, Jeong H, Yu M, Jeong J, Yu C. Development of sarcopenia assessment system using balance and gait ability:preliminary tests in the elderly. Technol Health Care 32: 447–455, 2024. doi:10.3233/THC-248039.
    Crossref | PubMed | Web of Science | Google Scholar
  • 966. Kim DH, Rockwood K. Frailty in older adults. N Engl J Med 391: 538–548, 2024. doi:10.1056/NEJMra2301292.
    Crossref | PubMed | Web of Science | Google Scholar
  • 967. Butler AA, Menant JC, Tiedemann AC, Lord SR. Age and gender differences in seven tests of functional mobility. J Neuroeng Rehabil 6: 31, 2009. doi:10.1186/1743-0003-6-31.
    Crossref | PubMed | Web of Science | Google Scholar
  • 968. Manca A, Fiorito G, Morrone M, Boi A, Mercante B, Martinez G, Ventura L, Delitala AP, Cano A, Catte MG, Solinas G, Melis F, Ginatempo F, Deriu F. A novel estimate of biological aging by multiple fitness tests is associated with risk scores for age-related diseases. Front Physiol 14: 1164943, 2023. doi:10.3389/fphys.2023.1164943.
    Crossref | PubMed | Web of Science | Google Scholar
  • 969. Taylor JA, Greenhaff PL, Bartlett DB, Jackson TA, Duggal NA, Lord JM. Multisystem physiological perspective of human frailty and its modulation by physical activity. Physiol Rev 103: 1137–1191, 2023. doi:10.1152/physrev.00037.2021.
    Link | Web of Science | Google Scholar
  • 970. Kwak D, Thompson LV. Frailty: past, present, and future? Sports Med Health Sci 3: 1–10, 2021. doi:10.1016/j.smhs.2020.11.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 971. Kojima G, Iliffe S, Walters K. Frailty index as a predictor of mortality: a systematic review and meta-analysis. Age Ageing 47: 193–200, 2018. doi:10.1093/ageing/afx162.
    Crossref | PubMed | Web of Science | Google Scholar
  • 972. de Fatima Ribeiro Silva C, Ohara DG, Matos AP, Pinto A, Pegorari MS. Short physical performance battery as a measure of physical performance and mortality predictor in older adults: a comprehensive literature review. Int J Environ Res Public Health 18: 10612, 2021. doi:10.3390/ijerph182010612.
    Crossref | PubMed | Web of Science | Google Scholar
  • 973. Woldemariam S, Oberndorfer M, Stein VK, Haider S, Dorner TE. Association between frailty and subsequent disability trajectories among older adults: a growth curve longitudinal analysis from the Survey of Health, Ageing and Retirement in Europe (2004-19). Eur J Public Health 34: 1184–1191, 2024. doi:10.1093/eurpub/ckae146.
    Crossref | PubMed | Web of Science | Google Scholar
  • 974. Stephan Y, Sutin AR, Luchetti M, Aschwanden D, Karakose S, Terracciano A. Balance, strength, and risk of dementia: findings from the Health and Retirement Study and the English Longitudinal Study of Ageing. J Gerontol A Biol Sci Med Sci 79: glae165, 2024. doi:10.1093/gerona/glae165.
    Crossref | PubMed | Web of Science | Google Scholar
  • 975. Löppönen A, Karavirta L, Finni T, Palmberg L, Portegijs E, Rantanen T, Delecluse C, Van Roie E, Rantalainen T. Free-living sit-to-stand characteristics as predictors of lower extremity functional decline among older adults. Med Sci Sports Exerc 56: 1672–1677, 2024. doi:10.1249/MSS.0000000000003470.
    Crossref | PubMed | Web of Science | Google Scholar
  • 976. Brito LB, Ricardo DR, Araújo DS, Ramos PS, Myers J, Araújo CG. Ability to sit and rise from the floor as a predictor of all-cause mortality. Eur J Prev Cardiol 21: 892–898, 2014. doi:10.1177/2047487312471759.
    Crossref | PubMed | Web of Science | Google Scholar
  • 977. Araujo CG, de Souza ES, Laukkanen JA, Fiatarone Singh M, Kunutsor SK, Myers J, Franca JF, Castro CL. Successful 10-second one-legged stance performance predicts survival in middle-aged and older individuals. Br J Sports Med 56: 975–980, 2022. doi:10.1136/bjsports-2021-105360.
    Crossref | PubMed | Web of Science | Google Scholar
  • 978. Tanaka T, Hase K, Mori K, Wakida M, Arima Y, Kubo T, Taguchi M. Stair-descent phenotypes in community-dwelling older adults determined using high-level balance tasks. Aging Clin Exp Res 37: 34, 2025. doi:10.1007/s40520-025-02929-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 979. de Souto Barreto P, Rolland Y, Ferrucci L, Arai H, Bischoff-Ferrari H, Duque G, Fielding RA, Beard JR, Muscedere J, Sierra F, Vellas B, LeBrasseur NK. Looking at frailty and intrinsic capacity through a geroscience lens: the ICFSR & Geroscience Task Force. Nat Aging 3: 1474–1479, 2023. doi:10.1038/s43587-023-00531-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 980. Mitnitski AB, Mogilner AJ, Rockwood K. Accumulation of deficits as a proxy measure of aging. ScientificWorldJournal 1: 323–336, 2001. doi:10.1100/tsw.2001.58.
    Crossref | PubMed | Google Scholar
  • 981. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Seeman T, Tracy R, Kop WJ, Burke G, McBurnie MA; Cardiovascular Health Study Collaborative Research Group. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 56: M146–M156, 2001. doi:10.1093/gerona/56.3.m146.
    Crossref | PubMed | Web of Science | Google Scholar
  • 982. Rose GA, Davies RG, Appadurai IR, Williams IM, Bashir M, Berg RM, Poole DC, Bailey DM. ‘Fit for surgery’: the relationship between cardiorespiratory fitness and postoperative outcomes. Exp Physiol 107: 787–799, 2022. doi:10.1113/EP090156.
    Crossref | PubMed | Web of Science | Google Scholar
  • 983. Wang H, Yang R, Xu J, Fang K, Abdelrahim M, Chang L. Sarcopenia as a predictor of postoperative risk of complications, mortality and length of stay following gastrointestinal oncological surgery. Ann R Coll Surg Engl 103: 630–637, 2021. doi:10.1308/rcsann.2021.0082.
    Crossref | PubMed | Web of Science | Google Scholar
  • 984. Yang Y, Sun M, Chen WM, Wu SY, Zhang J. Adverse postoperative outcomes in elderly patients with sarcopenia. BMC Geriatr 24: 561, 2024. doi:10.1186/s12877-024-05066-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 985. Afilalo J, Mottillo S, Eisenberg MJ, Alexander KP, Noiseux N, Perrault LP, Morin JF, Langlois Y, Ohayon SM, Monette J, Boivin JF, Shahian DM, Bergman H. Addition of frailty and disability to cardiac surgery risk scores identifies elderly patients at high risk of mortality or major morbidity. Circ Cardiovasc Qual Outcomes 5: 222–228, 2012. doi:10.1161/CIRCOUTCOMES.111.963157.
    Crossref | PubMed | Web of Science | Google Scholar
  • 986. Liu C, Li Y, Xu Y, Hou H. The impact of preoperative skeletal muscle mass index-defined sarcopenia on postoperative complications and survival in gastric cancer: an updated meta-analysis. Eur J Surg Oncol 51: 109569, 2025. doi:10.1016/j.ejso.2024.109569.
    Crossref | PubMed | Web of Science | Google Scholar
  • 987. Weerink LB, van Leeuwen BL, Kwee TC, Lamoth CJ, van Munster BC, de Bock GH. Co-occurrence of CT based radiological sarcopenia and frailty are related to impaired survival in surgical oncology. Br J Radiol 98: 607–613, 2025. doi:10.1093/bjr/tqaf023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 988. Butt JH, Petrie MC, Jhund PS, Sattar N, Desai AS, Køber L, Rouleau JL, Swedberg K, Zile MR, Solomon SD, Packer M, McMurray JJ. Anthropometric measures and adverse outcomes in heart failure with reduced ejection fraction: revisiting the obesity paradox. Eur Heart J 44: 1136–1153, 2023. doi:10.1093/eurheartj/ehad083.
    Crossref | PubMed | Web of Science | Google Scholar
  • 989. Lee SH, Jo J, Yang JH, Kim SM, Choi KH, Song YB, Jeong DS, Lee JM, Park TK, Hahn JY, Choi SH, Chung SR, Cho YH, Sung K, Kim WS, Gwon HC, Lee YT. Clinical impact of sarcopenia screening on long-term mortality in patients undergoing coronary bypass grafting. J Cachexia Sarcopenia Muscle 15: 2842–2851, 2024. doi:10.1002/jcsm.13645.
    Crossref | PubMed | Web of Science | Google Scholar
  • 990. Lees MJ, Prado CM, Wischmeyer PE, Phillips SM. Skeletal muscle: a critical organ for survival and recovery in critical illness. Crit Care Clin 41: 299–312, 2025. doi:10.1016/j.ccc.2024.08.011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 991. Umbrello M, Formenti P, Artale A, Assandri M, Palandri C, Ponti S, Venco R, Waccher G, Muttini S. Association between the ultrasound evaluation of muscle mass and adverse outcomes in critically ill patients: a prospective cohort study. Anesth Analg 140: 427–436, 2025. doi:10.1213/ANE.0000000000007035.
    Crossref | PubMed | Web of Science | Google Scholar
  • 992. Bernabei R, Landi F, Calvani R, Cesari M, Del Signore S, Anker SD, , et al.. Multicomponent intervention to prevent mobility disability in frail older adults: randomised controlled trial (SPRINTT project). BMJ 377: e068788, 2022. doi:10.1136/bmj-2021-068788.
    Crossref | PubMed | Web of Science | Google Scholar
  • 993. Valenzuela PL, Saco-Ledo G, Morales JS, Gallardo-Gómez D, Morales-Palomo F, López-Ortiz S, Rivas-Baeza B, Castillo-García A, Jiménez-Pavón D, Santos-Lozano A, Del Pozo Cruz B, Lucia A. Effects of physical exercise on physical function in older adults in residential care: a systematic review and network meta-analysis of randomised controlled trials. Lancet Healthy Longev 4: e247–e256, 2023. doi:10.1016/S2666-7568(23)00057-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 994. Zeng G, Lin Y, Xie P, Lin J, He Y, Wei J. Association between physical activity & sedentary time on frailty in adults with chronic kidney disease: cross-sectional NHANES study. Exp Gerontol 195: 112557, 2024. doi:10.1016/j.exger.2024.112557.
    Crossref | PubMed | Web of Science | Google Scholar
  • 995. Gill TM. Preserving community mobility in vulnerable older people. BMJ 377: o1084, 2022. doi:10.1136/bmj.o1084.
    Crossref | PubMed | Web of Science | Google Scholar
  • 996. Lavery JA, Boutros PC, Scott JM, Tammela T, Moskowitz CS, Jones LW. Pan-cancer analysis of postdiagnosis exercise and mortality. J Clin Oncol 41: 4982–4992, 2023. doi:10.1200/JCO.23.00058.
    Crossref | PubMed | Web of Science | Google Scholar
  • 997. Kanemura T, Takeoka T, Sugase T, Urakawa S, Masuike Y, Shinno N, Hara H, Kitakaze M, Kubo M, Mukai Y, Sueda T, Hasegawa S, Akita H, Nishimura J, Wada H, Yasui M, Omori T, Miyata H. Significance of comprehensive analysis of preoperative sarcopenia based on muscle mass, muscle strength, and physical function for the prognosis of patients with esophageal cancer. Ann Surg Oncol 31: 818–826, 2024. doi:10.1245/s10434-023-14306-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 998. Cannataro R, Cione E, Bonilla DA, Cerullo G, Angelini F, D’Antona G. Strength training in elderly: an useful tool against sarcopenia. Front Sports Act Living 4: 950949, 2022. doi:10.3389/fspor.2022.950949.
    Crossref | PubMed | Web of Science | Google Scholar
  • 999. Chun SY, Cho YS, Kim HB. Association between reduced muscle mass and poor prognosis of biliary sepsis. Sci Rep 14: 1857, 2024. doi:10.1038/s41598-024-52502-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1000. Markakis GE, Lai JC, Karakousis ND, Papatheodoridis GV, Psaltopoulou T, Merli M, Sergentanis TN, Cholongitas E. Sarcopenia as a predictor of survival and complications of patients with cirrhosis after liver transplantation: a systematic review and meta-analysis. Clin Transplant 39: e70088, 2025. doi:10.1111/ctr.70088.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1001. Brawner CA, Ehrman JK, Bole S, Kerrigan DJ, Parikh SS, Lewis BK, Gindi RM, Keteyian C, Abdul-Nour K, Keteyian SJ. Inverse relationship of maximal exercise capacity to hospitalization secondary to Coronavirus Disease 2019. Mayo Clin Proc 96: 32–39, 2021. doi:10.1016/j.mayocp.2020.10.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1002. Choi JY, Rhee CK, Kim SH, Jo YS. Muscle mass index decline as a predictor of lung function reduction in the general population. J Cachexia Sarcopenia Muscle 16: e13663, 2025. doi:10.1002/jcsm.13663.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1003. Veronese N, Soysal P, Demurtas J, Solmi M, Bruyère O, Christodoulou N, Ramalho R, Fusar-Poli P, Lappas AS, Pinto D, Frederiksen KS, Corbi GM, Karpenko O, Georges J, Durães J, Schlögl M, Yilmaz O, Sieber C, Shenkin SD, Smith L, Reginster JY, Maggi S, Limongi F, Ars J, Barbagallo M, Cherubini A, Quinn T; Alzheimer Europe, European College of Neuropsychopharmacology, European Geriatric Medicine Society, European Interdisciplinary Council on Ageing, European Society of Clinical and Economic Aspects of Osteoporosis and Osteoarthritis, International Association of Gerontology and Geriatrics-European Region, Scottish Brain Health ARC, World Psychiatry Association-Preventive Psychiatry Section, European Academy of Neurology. Physical activity and exercise for the prevention and management of mild cognitive impairment and dementia: a collaborative international guideline. Eur Geriatr Med 14: 925–952, 2023. doi:10.1007/s41999-023-00858-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1004. Soendenbroe C, Boraxbekk CJ, Mackey AL. Enhancing muscle and brain resilience: The role of prehabilitative exercise in mitigating disuse effects. J Physiol, 2025. doi:10.1113/JP284499.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1005. Prado CM, Phillips SM, Gonzalez MC, Heymsfield SB. Muscle matters: the effects of medically induced weight loss on skeletal muscle. Lancet Diabetes Endocrinol 12: 785–787, 2024. doi:10.1016/S2213-8587(24)00272-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1006. McCarthy D, Berg A. Weight loss strategies and the risk of skeletal muscle mass loss. Nutrients 13: 2473, 2021. doi:10.3390/nu13072473.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1007. Asher L, Aresu M, Falaschetti E, Mindell J. Most older pedestrians are unable to cross the road in time: a cross-sectional study. Age Ageing 41: 690–694, 2012. doi:10.1093/ageing/afs076.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1008. Seals DR, Justice JN, LaRocca TJ. Physiological geroscience: targeting function to increase healthspan and achieve optimal longevity. J Physiol 594: 2001–2024, 2016. doi:10.1113/jphysiol.2014.282665.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1009. Nolan CM, Kon SS, Patel S, Jones SE, Barker RE, Polkey MI, Maddocks M, Man WD. Gait speed and pedestrian crossings in COPD. Thorax 73: 191–192, 2018. doi:10.1136/thoraxjnl-2017-210173.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1010. Dommershuijsen LJ, Ragunathan J, Ruiter R, Groothof D, Mattace-Raso FU, Ikram MA, Polinder-Bos HA. Gait speed reference values in community-dwelling older adults—cross-sectional analysis from the Rotterdam Study. Exp Gerontol 158: 111646, 2022. doi:10.1016/j.exger.2021.111646.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1011. Hsu PS, Lee WJ, Peng LN, Lu WH, Meng LC, Hsiao FY, Chen LK. Safeguarding vitality and cognition: the role of sarcopenia in intrinsic capacity decline among octogenarians from multiple cohorts. J Nutr Health Aging 28: 100268, 2024. doi:10.1016/j.jnha.2024.100268.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1012. Rosas-Carrasco O, Manrique-Espinoza B, López-Alvarenga JC, Mena-Montes B, Omaña-Guzmán I. Osteosarcopenia predicts greater risk of functional disability than sarcopenia: a longitudinal analysis of FraDySMex cohort study. J Nutr Health Aging 28: 100368, 2024. doi:10.1016/j.jnha.2024.100368.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1013. Joy EA, Briesacher M, Wiegand B. Musculoskeletal failure. Am J Lifestyle Med 18: 826–829, 2024. doi:10.1177/15598276241256878.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1014. Izquierdo M, de Souto Barreto P, Arai H, Bischoff-Ferrari HA, Cadore EL, Cesari M, , et al.. Global consensus on optimal exercise recommendations for enhancing healthy longevity in older adults (ICFSR). J Nutr Health Aging 29: 100401, 2025. doi:10.1016/j.jnha.2024.100401.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1015. López-Ortiz S, Lista S, Valenzuela PL, Pinto-Fraga J, Carmona R, Caraci F, Caruso G, Toschi N, Emanuele E, Gabelle A, Nisticò R, Garaci F, Lucia A, Santos-Lozano A. Effects of physical activity and exercise interventions on Alzheimer’s disease: an umbrella review of existing meta-analyses. J Neurol 270: 711–725, 2023. doi:10.1007/s00415-022-11454-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1016. Cho Y, Jang H, Kwon S, Oh H. Aerobic, muscle-strengthening, and flexibility physical activity and risks of all-cause and cause-specific mortality: a population-based prospective cohort of Korean adults. BMC Public Health 23: 1148, 2023. doi:10.1186/s12889-023-15969-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1017. Araujo CG, de Souza ES, Kunutsor SK, Franklin BA, Laukkanen JA, Myers J, Fiatarone Singh MA, Franca JF, Castro CL. Reduced body flexibility is associated with poor survival in middle-aged men and women: a prospective cohort study. Scand J Med Sci Sports 34: e14708, 2024. doi:10.1111/sms.14708.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1018. Zhong YJ, Meng Q, Su CH. Mechanism-driven strategies for reducing fall risk in the elderly: a multidisciplinary review of exercise interventions. Healthcare (Basel) 12: 2394, 2024. doi:10.3390/healthcare12232394.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1019. Thompson WR, Sallis R, Joy E, Jaworski CA, Stuhr RM, Trilk JL. Exercise is medicine. Am J Lifestyle Med 14: 511–523, 2020. doi:10.1177/1559827620912192.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1020. Hedge ET, Brazile TL, Hughson RL, Levine BD. Plasticity of the heart in response to changes in physical activity. J Physiol, 2024. doi:10.1113/JP284158.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1021. Cavalcante BR, Falck RS, Liu-Ambrose T. Editorial: “may the force (and size) be with you”: muscle mass and function are important risk factors for cognitive decline and dementia. J Nutr Health Aging 27: 926–928, 2023. doi:10.1007/s12603-023-2023-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1022. Hu Y, Peng W, Ren R, Wang Y, Wang G. Sarcopenia and mild cognitive impairment among elderly adults: The first longitudinal evidence from CHARLS. J Cachexia Sarcopenia Muscle 13: 2944–2952, 2022. doi:10.1002/jcsm.13081.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1023. Peng TC, Chen WL, Wu LW, Chang YW, Kao TW. Sarcopenia and cognitive impairment: a systematic review and meta-analysis. Clin Nutr 39: 2695–2701, 2020. doi:10.1016/j.clnu.2019.12.014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1024. Liu M, He P, Ye Z, Zhang Y, Zhou C, Yang S, Zhang Y, Qin X. Association of handgrip strength and walking pace with incident Parkinson’s disease. J Cachexia Sarcopenia Muscle 15: 198–207, 2024. doi:10.1002/jcsm.13366.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1025. Jiang R, Westwater ML, Noble S, Rosenblatt M, Dai W, Qi S, Sui J, Calhoun VD, Scheinost D. Associations between grip strength, brain structure, and mental health in >40,000 participants from the UK Biobank. BMC Med 20: 286, 2022. doi:10.1186/s12916-022-02490-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1026. Zou L, Herold F, Cheval B, Wheeler MJ, Pindus DM, Erickson KI, Raichlen DA, Alexander GE, Müller NG, Dunstan DW, Kramer AF, Hillman CH, Hallgren M, Ekelund U, Maltagliati S, Owen N. Sedentary behavior and lifespan brain health. Trends Cogn Sci 28: 369–382, 2024. doi:10.1016/j.tics.2024.02.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1027. Iso-Markku P, Kujala UM, Knittle K, Polet J, Vuoksimaa E, Waller K. Physical activity as a protective factor for dementia and Alzheimer’s disease: systematic review, meta-analysis and quality assessment of cohort and case-control studies. Br J Sports Med 56: 701–709, 2022. doi:10.1136/bjsports-2021-104981.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1028. Min J, Cao Z, Duan T, Wang Y, Xu C. Accelerometer-derived ‘weekend warrior’ physical activity pattern and brain health. Nat Aging 4: 1394–1402, 2024. doi:10.1038/s43587-024-00688-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1029. Faulkner ME, Gong Z, Bilgel M, Laporte JP, Guo A, Bae J, Palchamy E, Kaileh M, Bergeron CM, Bergeron J, Church S, D’Agostino J, Ferrucci L, Bouhrara M. Evidence of association between higher cardiorespiratory fitness and higher cerebral myelination in aging. Proc Natl Acad Sci USA 121: e2402813121, 2024. doi:10.1073/pnas.2402813121.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1030. Zhang W, Zhou C, Chen A. A systematic review and meta-analysis of the effects of physical exercise on white matter integrity and cognitive function in older adults. Geroscience 46: 2641–2651, 2024. doi:10.1007/s11357-023-01033-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1031. Marseglia A, Dartora C, Samuelsson J, Poulakis K, Mohanty R, Shams S, Lindberg O, Rydén L, Sterner TR, Skoog J, Zettergren A, Kern S, Skoog I, Westman E. Biological brain age and resilience in cognitively unimpaired 70-year-old individuals. Alzheimers Dement 21: e14435, 2025. doi:10.1002/alz.14435.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1032. Samuelsson J, Marseglia A, Wallengren O, Lindberg O, Dartora C, Cedres N, Shams S, Kern S, Zettergren A, Westman E, Skoog I. Association of body composition with neuroimaging biomarkers and cognitive function; a population-based study of 70-year-olds. EBioMedicine 112: 105555, 2025. doi:10.1016/j.ebiom.2024.105555.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1033. Anstey KJ. Total physical activity matters for brain health. Nat Aging 4: 1340–1342, 2024. doi:10.1038/s43587-024-00712-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1034. O’Donovan G, Lee IM, Hamer M, Stamatakis E. Association of “weekend warrior” and other leisure time physical activity patterns with risks for all-cause, cardiovascular disease, and cancer mortality. JAMA Intern Med 177: 335–342, 2017. doi:10.1001/jamainternmed.2016.8014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1035. Shiroma EJ, Lee IM, Schepps MA, Kamada M, Harris TB. Physical activity patterns and mortality: the weekend warrior and activity bouts. Med Sci Sports Exerc 51: 35–40, 2019. doi:10.1249/MSS.0000000000001762.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1036. Khurshid S, Al-Alusi MA, Churchill TW, Guseh JS, Ellinor PT. Accelerometer-derived “weekend warrior” physical activity and incident cardiovascular disease. JAMA 330: 247–252, 2023. doi:10.1001/jama.2023.10875.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1037. Boa Sorte Silva NC, Barha CK, Erickson KI, Kramer AF, Liu-Ambrose T. Physical exercise, cognition, and brain health in aging. Trends Neurosci 47: 402–417, 2024. doi:10.1016/j.tins.2024.04.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1038. Steves CJ, Mehta MM, Jackson SH, Spector TD. Kicking back cognitive ageing: leg power predicts cognitive ageing after ten years in older female twins. Gerontology 62: 138–149, 2016. doi:10.1159/000441029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1039. Nyberg J, Åberg MA, Schiöler L, Nilsson M, Wallin A, Torén K, Kuhn HG. Cardiovascular and cognitive fitness at age 18 and risk of early-onset dementia. Brain 137: 1514–1523, 2014. doi:10.1093/brain/awu041.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1040. Hörder H, Johansson L, Guo X, Grimby G, Kern S, Östling S, Skoog I. Midlife cardiovascular fitness and dementia: a 44-year longitudinal population study in women. Neurology 90: e1298–e1305, 2018. doi:10.1212/WNL.0000000000005290.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1041. Brandhorst S, Longo VD. Exploring juventology: unlocking the secrets of youthspan and longevity programs. Front Aging 5: 1379289, 2024. doi:10.3389/fragi.2024.1379289.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1042. Belsky DW, Caspi A, Houts R, Cohen HJ, Corcoran DL, Danese A, Harrington H, Israel S, Levine ME, Schaefer JD, Sugden K, Williams B, Yashin AI, Poulton R, Moffitt TE. Quantification of biological aging in young adults. Proc Natl Acad Sci USA 112: E4104–E4110, 2015. doi:10.1073/pnas.1506264112.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1043. Furrer R, Heim B, Schmid S, Dilbaz S, Adak V, Nordström KJ, Ritz D, Steurer SA, Walter J, Handschin C. Molecular control of endurance training adaptation in male mouse skeletal muscle. Nat Metab 5: 2020–2035, 2023. doi:10.1038/s42255-023-00891-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1044. Koch LG, Kemi OJ, Qi N, Leng SX, Bijma P, Gilligan LJ, Wilkinson JE, Wisløff H, Høydal MA, Rolim N, Abadir PM, van Grevenhof EM, Smith GL, Burant CF, Ellingsen O, Britton SL, Wisløff U. Intrinsic aerobic capacity sets a divide for aging and longevity. Circ Res 109: 1162–1172, 2011. doi:10.1161/CIRCRESAHA.111.253807.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1045. Battilana F, Steurer S, Rizzi G, Delgado AC, Tan KR, Handschin C. Exercise-linked improvement in age-associated loss of balance is associated with increased vestibular input to motor neurons. Aging Cell 19: e13274, 2020. doi:10.1111/acel.13274.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1046. Fuller KN, Thyfault JP. Barriers in translating preclinical rodent exercise metabolism findings to human health. J Appl Physiol (1985) 130: 182–192, 2021. doi:10.1152/japplphysiol.00683.2020.
    Link | Web of Science | Google Scholar
  • 1047. Chen Z, Raj A, Prateek GV, Di Francesco A, Liu J, Keyes BE, Kolumam G, Jojic V, Freund A. Automated, high-dimensional evaluation of physiological aging and resilience in outbred mice. eLife 11: e72664, 2022. doi:10.7554/eLife.72664.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1048. Dennison EM, Laskou F, Westbury LD, Bevilacqua G, Fuggle NR, Iidaka T, Horii C, Tanaka S, Yoshimura N, Cooper C. Do lifestyle, anthropometric and demographic factors associated with muscle strength differ in a UK and Japanese cohort? An exploratory analysis. Aging Clin Exp Res 35: 3097–3104, 2023. doi:10.1007/s40520-023-02614-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1049. Rantanen T, Masaki K, He Q, Ross GW, Willcox BJ, White L. Midlife muscle strength and human longevity up to age 100 years: a 44-year prospective study among a decedent cohort. Age (Dordr) 34: 563–570, 2012. doi:10.1007/s11357-011-9256-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1050. Onerup A, Mehlig K, Geijerstam AA, Ekblom-Bak E, Kuhn HG, Lissner L, Åberg M, Börjesson M. Associations between cardiorespiratory fitness in youth and the incidence of site-specific cancer in men: a cohort study with register linkage. Br J Sports Med 57: 1248–1256, 2023. doi:10.1136/bjsports-2022-106617.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1051. Teraž K, Kalc M, Šimunic B, Marusic U, Pori P, Pišot S, Pišot R. Participation in youth sports influences sarcopenia parameters in older adults. PeerJ 11: e16432, 2023. doi:10.7717/peerj.16432.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1052. Lesinski M, Herz M, Schmelcher A, Granacher U. Effects of resistance training on physical fitness in healthy children and adolescents: an umbrella review. Sports Med 50: 1901–1928, 2020. doi:10.1007/s40279-020-01327-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1053. Hetherington-Rauth M, Magalhães JP, Alcazar J, Rosa GB, Correia IR, Ara I, Sardinha LB. Relative sit-to-stand muscle power predicts an older adult’s physical independence at age of 90 yrs beyond that of relative handgrip strength, physical activity, and sedentary time: a cross-sectional analysis. Am J Phys Med Rehabil 101: 995–1000, 2022. doi:10.1097/PHM.0000000000001945.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1054. Manning KM, Hall KS, Sloane R, Magistro D, Rabaglietti E, Lee CC, Castle S, Kopp T, Giffuni J, Katzel L, McDonald M, Miyamoto M, Pearson M, Jennings SC, Bettger JP, Morey MC. Longitudinal analysis of physical function in older adults: the effects of physical inactivity and exercise training. Aging Cell 23: e13987, 2024. doi:10.1111/acel.13987.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1055. Tiedemann A, Sherrington C, Lord SR. Physiological and psychological predictors of walking speed in older community-dwelling people. Gerontology 51: 390–395, 2005. doi:10.1159/000088703.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1056. Marzuca-Nassr GN, Alegría-Molina A, SanMartín-Calísto Y, Artigas-Arias M, Huard N, Sapunar J, Salazar LA, Verdijk LB, van Loon LJ. Muscle mass and strength gains following resistance exercise training in older adults 65-75 years and older adults above 85 years. Int J Sport Nutr Exerc Metab 34: 11–19, 2024. doi:10.1123/ijsnem.2023-0087.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1057. Mende E, Moeinnia N, Schaller N, Weiß M, Haller B, Halle M, Siegrist M. Progressive machine-based resistance training for prevention and treatment of sarcopenia in the oldest old: a systematic review and meta-analysis. Exp Gerontol 163: 111767, 2022. doi:10.1016/j.exger.2022.111767.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1058. Carrick-Ranson G, Howden EJ, Levine BD. Exercise in octogenarians: how much is too little? Annu Rev Med 73: 377–391, 2022. doi:10.1146/annurev-med-070119-115343.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1059. Fisher J, Steele J, McKinnon P, McKinnon S. Strength gains as a result of brief, infrequent resistance exercise in older adults. J Sports Med 2014: 731890, 2014. doi:10.1155/2014/731890.
    Crossref | PubMed | Google Scholar
  • 1060. The Lancet Healthy Longevity. Physical activity knows no age limit. Lancet Healthy Longev 5: e447, 2024. doi:10.1016/S2666-7568(24)00115-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1061. McKendry J, Breen L, Shad BJ, Greig CA. Muscle morphology and performance in master athletes: a systematic review and meta-analyses. Ageing Res Rev 45: 62–82, 2018. doi:10.1016/j.arr.2018.04.007.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1062. Fiatarone MA, Marks EC, Ryan ND, Meredith CN, Lipsitz LA, Evans WJ. High-intensity strength training in nonagenarians. Effects on skeletal muscle. JAMA 263: 3029–3034, 1990. doi:10.1001/jama.1990.03440220053029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1063. de Santana DA, Scolfaro PG, Marzetti E, Cavaglieri CR. Lower extremity muscle hypertrophy in response to resistance training in older adults: systematic review, meta-analysis, and meta-regression of randomized controlled trials. Exp Gerontol 198: 112639, 2024. doi:10.1016/j.exger.2024.112639.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1064. Fuchs CJ, Trommelen J, Weijzen ME, Smeets JS, van Kranenburg J, Verdijk LB, van Loon LJ. Becoming a world champion powerlifter at 71 years of age: it is never too late to start exercising. Int J Sport Nutr Exerc Metab 34: 223–231, 2024. doi:10.1123/ijsnem.2023-0230.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1065. Lavisci P, Myers J, Grazzi G. Exceptional exercise capacity in a late bloomer octogenarian triathlete. J Sports Med Phys Fitness 65: 90–94, 2025. doi:10.23736/S0022-4707.24.15979-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1066. Moore SC, Patel AV, Matthews CE, Berrington de Gonzalez A, Park Y, Katki HA, Linet MS, Weiderpass E, Visvanathan K, Helzlsouer KJ, Thun M, Gapstur SM, Hartge P, Lee IM. Leisure time physical activity of moderate to vigorous intensity and mortality: a large pooled cohort analysis. PLoS Med 9: e1001335, 2012. doi:10.1371/journal.pmed.1001335.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1067. López-Bueno R, Yang L, Stamatakis E, Del Pozo Cruz B. Moderate and vigorous leisure time physical activity in older adults and Alzheimer’s disease-related mortality in the USA: a dose-response, population-based study. Lancet Healthy Longev 4: e703–e710, 2023. doi:10.1016/S2666-7568(23)00212-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1068. Raj V, Stogios N, Agarwal SM, Cheng AJ. The neuromuscular basis of functional impairment in schizophrenia: a scoping review. Schizophr Res 274: 46–56, 2024. doi:10.1016/j.schres.2024.09.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1069. Chen LJ, Hamer M, Lai YJ, Huang BH, Ku PW, Stamatakis E. Can physical activity eliminate the mortality risk associated with poor sleep? A 15-year follow-up of 341,248 MJ Cohort participants. J Sport Health Sci 11: 596–604, 2022. doi:10.1016/j.jshs.2021.03.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1070. Chastin S, McGregor D, Palarea-Albaladejo J, Diaz KM, Hagströmer M, Hallal PC, van Hees VT, Hooker S, Howard VJ, Lee IM, von Rosen P, Sabia S, Shiroma EJ, Yerramalla MS, Dall P. Joint association between accelerometry-measured daily combination of time spent in physical activity, sedentary behaviour and sleep and all-cause mortality: a pooled analysis of six prospective cohorts using compositional analysis. Br J Sports Med 55: 1277–1285, 2021. doi:10.1136/bjsports-2020-102345.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1071. Cadore EL, Izquierdo M, Teodoro JL, Martínez-Velilla N, Zambom-Ferraresi F, Moriguchi EH, Saez de Asteasu ML. Effects of short-term multicomponent exercise intervention on muscle power in hospitalized older patients: a secondary analysis of a randomized clinical trial. J Cachexia Sarcopenia Muscle 14: 2959–2968, 2023. doi:10.1002/jcsm.13375.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1072. Bilberg A, Mannerkorpi K, Borjesson M, Svedlund S, Sivertsson J, Klingberg E, Bjersing J. High-intensity interval training improves cardiovascular and physical health in patients with rheumatoid arthritis: a multicentre randomised controlled trial. Br J Sports Med 58: 1409–1418, 2024. doi:10.1136/bjsports-2024-108369.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1073. Rossi A, Dikareva A, Bacon SL, Daskalopoulou SS. The impact of physical activity on mortality in patients with high blood pressure: a systematic review. J Hypertens 30: 1277–1288, 2012. doi:10.1097/HJH.0b013e3283544669.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1074. Cornelissen VA, Fagard RH. Effects of endurance training on blood pressure, blood pressure-regulating mechanisms, and cardiovascular risk factors. Hypertension 46: 667–675, 2005. doi:10.1161/01.HYP.0000184225.05629.51.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1075. Cornelissen VA, Smart NA. Exercise training for blood pressure: a systematic review and meta-analysis. J Am Heart Assoc 2: e004473, 2013. doi:10.1161/JAHA.112.004473.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1076. La Gerche A. Proof that exercise works, now it’s time for optimizing delivery to our patients with pulmonary hypertension. Eur Heart J 42: 2296–2298, 2021. doi:10.1093/eurheartj/ehaa780.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1077. Grünig E, MacKenzie A, Peacock AJ, Eichstaedt CA, Benjamin N, Nechwatal R, , et al.. Standardized exercise training is feasible, safe, and effective in pulmonary arterial and chronic thromboembolic pulmonary hypertension: results from a large European multicentre randomized controlled trial. Eur Heart J 42: 2284–2295, 2021. doi:10.1093/eurheartj/ehaa696.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1078. Mourtzakis M, Heckman GA, McKelvie RS. Aging with heart failure: muscle matters. Can J Cardiol 40: 2552–2554, 2024. doi:10.1016/j.cjca.2024.09.032.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1079. Jansen J, Marshall PW, Benatar JR, Cross R, Lindbom TK, Kingsley M. Low-intensity resistance exercise in cardiac rehabilitation: a narrative review of mechanistic evidence and clinical implications. J Clin Med 13: 7338, 2024. doi:10.3390/jcm13237338.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1080. Watz H, Pitta F, Rochester CL, Garcia-Aymerich J, ZuWallack R, Troosters T, Vaes AW, Puhan MA, Jehn M, Polkey MI, Vogiatzis I, Clini EM, Toth M, Gimeno-Santos E, Waschki B, Esteban C, Hayot M, Casaburi R, Porszasz J, McAuley E, Singh SJ, Langer D, Wouters EF, Magnussen H, Spruit MA. An official European Respiratory Society statement on physical activity in COPD. Eur Respir J 44: 1521–1537, 2014. doi:10.1183/09031936.00046814.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1081. Spruit MA, Singh SJ, Garvey C, ZuWallack R, Nici L, Rochester C, , et al.. An official American Thoracic Society/European Respiratory Society statement: key concepts and advances in pulmonary rehabilitation. Am J Respir Crit Care Med 188: e13–e64, 2013. doi:10.1164/rccm.201309-1634ST.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1082. Garcia-Aymerich J, Lange P, Benet M, Schnohr P, Antó JM. Regular physical activity reduces hospital admission and mortality in chronic obstructive pulmonary disease: a population based cohort study. Thorax 61: 772–778, 2006. doi:10.1136/thx.2006.060145.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1083. Dai J, Dai W, Li WQ. Association of muscle-strengthening and aerobic physical activity with all-cause, cardiovascular disease, and cancer mortality in U.S. adults with diabetes. Eur J Prev Cardiol 2025: zwaf061, 2025. doi:10.1093/eurjpc/zwaf061.
    Crossref | Web of Science | Google Scholar
  • 1084. Qian C, Zhou F, Lu D, Huang J, Sun M. Exercise intensity and mortality in overweight and obese patients with chronic kidney disease: longitudinal analysis (1999–2016). BMC Public Health 24: 3020, 2024. doi:10.1186/s12889-024-20498-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1085. Zhang F, Wang H, Bai Y, Huang L, Zhong Y, Li Y. Gait speed and all-cause mortality in whole-spectrum chronic kidney disease: a systematic review and meta-analysis included 6217 participants. J Cachexia Sarcopenia Muscle 16: e13739, 2025. doi:10.1002/jcsm.13739.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1086. Tian S, Liang Z, Qui F, Yu Y, Wang C, Zhang M, Wang X. Optimal exercise modality and dose to improve depressive symptoms in adults with major depressive disorder: a systematic review and Bayesian model-based network meta-analysis of RCTs. J Psychiatr Res 176: 384–392, 2024. doi:10.1016/j.jpsychires.2024.06.031.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1087. O’Neil A, Perez J, Young LM, John T, Turner M, Saunders D, , et al.. Clinical and cost-effectiveness of remote-delivered, online lifestyle therapy versus psychotherapy for reducing depression: results from the CALM non-inferiority, randomised trial. Lancet Reg Health West Pac 49: 101142, 2024. doi:10.1016/j.lanwpc.2024.101142.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1088. Veronese N, Ragusa FS, Hajek A, Stubbs B, Smith L, Barbagallo M, Dominguez LJ, Fontana L, Soysal P, Sabico S, Al-Daghri NM. Long-term impact of physical activity on mortality in adults with multimorbidity: a 12-year cohort longitudinal study from the survey on health, ageing and retirement in Europe. J Cachexia Sarcopenia Muscle 16: e13695, 2025. doi:10.1002/jcsm.13695.
    Crossref | Web of Science | Google Scholar
  • 1089. Vøllestad NK, Mengshoel AM. Post-exertional malaise in daily life and experimental exercise models in patients with myalgic encephalomyelitis/chronic fatigue syndrome. Front Physiol 14: 1257557, 2023. doi:10.3389/fphys.2023.1257557.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1090. Paffenbarger RS Jr, Hyde RT, Wing AL, Hsieh CC. Physical activity, all-cause mortality, and longevity of college alumni. N Engl J Med 314: 605–613, 1986. doi:10.1056/NEJM198603063141003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1091. Paffenbarger RS Jr, Hyde RT, Wing AL, Lee IM, Jung DL, Kampert JB. The association of changes in physical-activity level and other lifestyle characteristics with mortality among men. N Engl J Med 328: 538–545, 1993. doi:10.1056/NEJM199302253280804.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1092. Kujala UM, Leskinen T, Rottensteiner M, Aaltonen S, Ala-Korpela M, Waller K, Kaprio J. Physical activity and health: findings from Finnish monozygotic twin pairs discordant for physical activity. Scand J Med Sci Sports 32: 1316–1323, 2022. doi:10.1111/sms.14205.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1093. Berntzen B, Jukarainen S, Kataja M, Hakkarainen A, Lundbom J, Lundbom N, Tammelin T, Simonen R, Piirilä P, Rissanen A, Kaprio J, Paavonen EJ, Pietiläinen KH. Physical activity, cardiorespiratory fitness, and metabolic outcomes in monozygotic twin pairs discordant for body mass index. Scand J Med Sci Sports 28: 1048–1055, 2018. doi:10.1111/sms.12975.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1094. Bathgate KE, Bagley JR, Jo E, Talmadge RJ, Tobias IS, Brown LE, Coburn JW, Arevalo JA, Segal NL, Galpin AJ. Muscle health and performance in monozygotic twins with 30 years of discordant exercise habits. Eur J Appl Physiol 118: 2097–2110, 2018. doi:10.1007/s00421-018-3943-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1095. Kujala UM, Kaprio J, Sarna S, Koskenvuo M. Relationship of leisure-time physical activity and mortality: the Finnish twin cohort. JAMA 279: 440–444, 1998. doi:10.1001/jama.279.6.440.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1096. Churchward-Venne TA, Tieland M, Verdijk LB, Leenders M, Dirks ML, de Groot LC, van Loon LJ. There are no nonresponders to resistance-type exercise training in older men and women. J Am Med Dir Assoc 16: 400–411, 2015. doi:10.1016/j.jamda.2015.01.071.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1097. Atkinson G, Batterham AM. True and false interindividual differences in the physiological response to an intervention. Exp Physiol 100: 577–588, 2015. doi:10.1113/EP085070.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1098. Renwick JR, Preobrazenski N, Wu Z, Khansari A, LeBouedec MA, Nuttall JM, Bancroft KR, Simpson-Stairs N, Swinton PA, Gurd BJ. Standard deviation of individual response for VO2max following exercise interventions: a systematic review and meta-analysis. Sports Med 54: 3069–3080, 2024. doi:10.1007/s40279-024-02089-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1099. Steele J, Fisher JP, Smith D, Schoenfeld BJ, Yang Y, Nakagawa S. Meta-analysis of variation in sport and exercise science: examples of application within resistance training research. J Sports Sci 41: 1617–1634, 2023. doi:10.1080/02640414.2023.2286748.
    Crossref | PubMed | Google Scholar
  • 1100. Ross R, de Lannoy L, Stotz PJ. Separate effects of intensity and amount of exercise on interindividual cardiorespiratory fitness response. Mayo Clin Proc 90: 1506–1514, 2015. doi:10.1016/j.mayocp.2015.07.024.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1101. Austad SN, Bartke A. Sex differences in longevity and in responses to anti-aging interventions: a mini-review. Gerontology 62: 40–46, 2016. doi:10.1159/000381472.
    Crossref | Web of Science | Google Scholar
  • 1102. Kane AE, Howlett SE. Sex differences in frailty: comparisons between humans and preclinical models. Mech Ageing Dev 198: 111546, 2021. doi:10.1016/j.mad.2021.111546.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1103. Zeidan RS, McElroy T, Rathor L, Martenson MS, Lin Y, Mankowski RT. Sex differences in frailty among older adults. Exp Gerontol 184: 112333, 2023. doi:10.1016/j.exger.2023.112333.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1104. Gordon EH, Peel NM, Samanta M, Theou O, Howlett SE, Hubbard RE. Sex differences in frailty: a systematic review and meta-analysis. Exp Gerontol 89: 30–40, 2017. doi:10.1016/j.exger.2016.12.021.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1105. O’Caoimh R, Sezgin D, O'Donovan MR, Molloy DW, Clegg A, Rockwood K, Liew A. Prevalence of frailty in 62 countries across the world: a systematic review and meta-analysis of population-level studies. Age Ageing 50: 96–104, 2021. doi:10.1093/ageing/afaa219.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1106. Hubbard RE, Rockwood K. Frailty in older women. Maturitas 69: 203–207, 2011. doi:10.1016/j.maturitas.2011.04.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1107. Park C, Ko FC. The science of frailty: sex differences. Clin Geriatr Med 37: 625–638, 2021. doi:10.1016/j.cger.2021.05.008.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1108. Gordon EH, Hubbard RE. Do sex differences in chronic disease underpin the sex-frailty paradox? Mech Ageing Dev 179: 44–50, 2019. doi:10.1016/j.mad.2019.02.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1109. Hunter SK, Senefeld JW. Sex differences in human performance. J Physiol 602: 4129–4156, 2024. doi:10.1113/JP284198.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1110. Burns SD, Ailshire JA, Crimmins EM. Functional limitation among middle age and older adults: exploring cross-national gender disparities. Arch Gerontol Geriatr 123: 105410, 2024. doi:10.1016/j.archger.2024.105410.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1111. Atance D, Claramunt MM, Varea X, Aburto JM. Convergence and divergence in mortality: a global study from 1990 to 2030. PLoS One 19: e0295842, 2024. doi:10.1371/journal.pone.0295842.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1112. Carmel S. Health and well-being in late life: gender differences worldwide. Front Med (Lausanne) 6: 218, 2019. doi:10.3389/fmed.2019.00218.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1113. Ji H, Gulati M, Huang TY, Kwan AC, Ouyang D, Ebinger JE, Casaletto K, Moreau KL, Skali H, Cheng S. Sex differences in association of physical activity with all-cause and cardiovascular mortality. J Am Coll Cardiol 83: 783–793, 2024. doi:10.1016/j.jacc.2023.12.019.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1114. Puett D. Biology of aging: identified drivers and interventions for optimal healthspan. ACSM Health Fit J 22: 17–27, 2018. doi:10.1249/FIT.0000000000000360.
    Crossref | Web of Science | Google Scholar
  • 1115. Littlejohns TJ, Holliday J, Gibson LM, Garratt S, Oesingmann N, Alfaro-Almagro F, Bell JD, Boultwood C, Collins R, Conroy MC, Crabtree N, Doherty N, Frangi AF, Harvey NC, Leeson P, Miller KL, Neubauer S, Petersen SE, Sellors J, Sheard S, Smith SM, Sudlow CL, Matthews PM, Allen NE. The UK Biobank imaging enhancement of 100,000 participants: rationale, data collection, management and future directions. Nat Commun 11: 2624, 2020. doi:10.1038/s41467-020-15948-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1116. Bamberg F, Kauczor HU, Weckbach S, Schlett CL, Forsting M, Ladd SC, , et al.. Whole-body MR imaging in the German National Cohort: rationale, design, and technical background. Radiology 277: 206–220, 2015. doi:10.1148/radiol.2015142272.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1117. Lavie CJ, Arena R, Kaminsky LA. Making the case to measure and improve cardiorespiratory fitness in routine clinical practice. Mayo Clin Proc 97: 1038–1040, 2022. doi:10.1016/j.mayocp.2022.04.011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1118. Kaminsky LA, Imboden MT, Ozemek C. It’s time to (again) recognize the considerable clinical and public health significance of cardiorespiratory fitness. J Am Coll Cardiol 81: 1148–1150, 2023. doi:10.1016/j.jacc.2023.02.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1119. Ross R, Arena R, Myers J, Kokkinos P, Kaminsky LA. Update to the 2016 American Heart Association cardiorespiratory fitness statement. Prog Cardiovasc Dis 83: 10–15, 2024. doi:10.1016/j.pcad.2024.02.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1120. Kaminsky LA, Arena R, Myers J, Peterman JE, Bonikowske AR, Harber MP, Medina Inojosa JR, Lavie CJ, Squires RW. Updated reference standards for cardiorespiratory fitness measured with cardiopulmonary exercise testing: data from the Fitness Registry and the Importance of Exercise National Database (FRIEND). Mayo Clin Proc 97: 285–293, 2022. doi:10.1016/j.mayocp.2021.08.020.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1121. Peterman JE, Arena R, Myers J, Marzolini S, Ross R, Lavie CJ, Wisløff U, Stensvold D, Kaminsky LA. Development of global reference standards for directly measured cardiorespiratory fitness: a report from the Fitness Registry and Importance of Exercise National Database (FRIEND). Mayo Clin Proc 95: 255–264, 2020. doi:10.1016/j.mayocp.2019.06.013.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1122. Ross R, Blair SN, Arena R, Church TS, Despres JP, Franklin BA, Haskell WL, Kaminsky LA, Levine BD, Lavie CJ, Myers J, Niebauer J, Sallis R, Sawada SS, Sui X, Wisløff U; American Heart Association Physical Activity Committee of the Council on Lifestyle and Cardiometabolic Health, Council on Clinical Cardiology, Council on Epidemiology and Prevention, Council on Cardiovascular and Stroke Nursing, Council on Functional Genomics and Translational Biology, Stroke Council. Importance of Assessing Cardiorespiratory Fitness in Clinical Practice: A Case for Fitness as a Clinical Vital Sign: a scientific statement from the American Heart Association. Circulation 134: e653–e699, 2016. doi:10.1161/CIR.0000000000000461.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1123. Guazzi M, Arena R, Halle M, Piepoli MF, Myers J, Lavie CJ. 2016 focused update: clinical recommendations for cardiopulmonary exercise testing data assessment in specific patient populations. Circulation 133: e694–e711, 2016. doi:10.1161/CIR.0000000000000406.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1124. Guazzi M, Arena R, Halle M, Piepoli MF, Myers J, Lavie CJ. 2016 focused update: clinical recommendations for cardiopulmonary exercise testing data assessment in specific patient populations. Eur Heart J 39: 1144–1161, 2018. doi:10.1093/eurheartj/ehw180.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1125. Price OJ, Tsakirides C, Gray M, Stavropoulos-Kalinoglou A. ACSM Preparticipation Health Screening Guidelines: a UK University Cohort perspective. Med Sci Sports Exerc 51: 1047–1054, 2019. doi:10.1249/MSS.0000000000001868.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1126. Noonan V, Dean E. Submaximal exercise testing: clinical application and interpretation. Phys Ther 80: 782–807, 2000. doi:10.1093/ptj/80.8.782.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1127. Lobelo F, Rohm Young D, Sallis R, Garber MD, Billinger SA, Duperly J, Hutber A, Pate RR, Thomas RJ, Widlansky ME, McConnell MV, Joy EA; American Heart Association Physical Activity Committee of the Council on Lifestyle and Cardiometabolic Health, Council on Epidemiology and Prevention, Council on Clinical Cardiology, Council on Genomic and Precision Medicine, Council on Cardiovascular Surgery and Anesthesia, Stroke Council. Routine Assessment and Promotion of Physical Activity in Healthcare Settings: a scientific statement from the American Heart Association. Circulation 137: e495–e522, 2018. doi:10.1161/CIR.0000000000000559.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1128. Maron BJ, Levine BD, Washington RL, Baggish AL, Kovacs RJ, Maron MS; American Heart Association Echocardiography and Arrhythmias Committee of Council on Clinical Cardiology, Council on Cardiovascular Disease in Young, Council on Cardiovascular and Stroke Nursing, Council on Functional Genomics and Translational Biology, and American College of Cardiology. Eligibility and Disqualification Recommendations for Competitive Athletes With Cardiovascular Abnormalities: Task Force 2: Preparticipation Screening for Cardiovascular Disease in Competitive Athletes: a scientific statement from the American Heart Association and American College of Cardiology. Circulation 132: e267–e272, 2015. doi:10.1161/CIR.0000000000000238.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1129. Wolf C, Blackwell TL, Johnson E, Glynn NW, Nicklas B, Kritchevsky SB, Carnero EA, Cawthon PM, Cummings SR, Toledo FGS, Newman AB, Forman DE, Goodpaster BH. Cardiopulmonary exercise testing in a prospective multicenter cohort of older adults. Med Sci Sports Exerc 56: 1574–1584, 2024. doi:10.1249/MSS.0000000000003444.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1130. Albouaini K, Egred M, Alahmar A, Wright DJ. Cardiopulmonary exercise testing and its application. Postgrad Med J 83: 675–682, 2007. doi:10.1136/hrt.2007.121558.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1131. Ekblom-Bak E, Björkman F, Hellenius ML, Ekblom B. A new submaximal cycle ergometer test for prediction of VO2max. Scand J Med Sci Sports 24: 319–326, 2014. doi:10.1111/sms.12014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1132. Lee DC, Lavie CJ, Sui X, Blair SN. Running and mortality: is more actually worse? Mayo Clin Proc 91: 534–536, 2016. doi:10.1016/j.mayocp.2016.01.013.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1133. Mozaffarian D, Furberg CD, Psaty BM, Siscovick D. Physical activity and incidence of atrial fibrillation in older adults: the cardiovascular health study. Circulation 118: 800–807, 2008. doi:10.1161/CIRCULATIONAHA.108.785626.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1134. Abdulla J, Nielsen JR. Is the risk of atrial fibrillation higher in athletes than in the general population? A systematic review and meta-analysis. Europace 11: 1156–1159, 2009. doi:10.1093/europace/eup197.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1135. Qureshi WT, Alirhayim Z, Blaha MJ, Juraschek SP, Keteyian SJ, Brawner CA, Al-Mallah MH. Cardiorespiratory fitness and risk of incident atrial fibrillation: results from the Henry Ford Exercise Testing (FIT) Project. Circulation 131: 1827–1834, 2015. doi:10.1161/CIRCULATIONAHA.114.014833.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1136. Faselis C, Kokkinos P, Tsimploulis A, Pittaras A, Myers J, Lavie CJ, Kyritsi F, Lovic D, Karasik P, Moore H. Exercise capacity and atrial fibrillation risk in veterans: a cohort study. Mayo Clin Proc 91: 558–566, 2016. doi:10.1016/j.mayocp.2016.03.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1137. Arem H, Moore SC, Patel A, Hartge P, Berrington de Gonzalez A, Visvanathan K, Campbell PT, Freedman M, Weiderpass E, Adami HO, Linet MS, Lee IM, Matthews CE. Leisure time physical activity and mortality: a detailed pooled analysis of the dose-response relationship. JAMA Intern Med 175: 959–967, 2015. doi:10.1001/jamainternmed.2015.0533.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1138. Schnohr P, O'Keefe JH, Lavie CJ, Holtermann A, Lange P, Jensen GB, Marott JL. U-shaped association between duration of sports activities and mortality: Copenhagen City Heart Study. Mayo Clin Proc 96: 3012–3020, 2021. doi:10.1016/j.mayocp.2021.05.028.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1139. Schnohr P, O’Keefe JH, Marott JL, Lange P, Jensen GB. Dose of jogging and long-term mortality: the Copenhagen City Heart Study. J Am Coll Cardiol 65: 411–419, 2015. doi:10.1016/j.jacc.2014.11.023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1140. De Bosscher R, Dausin C, Claus P, Bogaert J, Dymarkowski S, Goetschalckx K, Ghekiere O, Belmans A, Van De Heyning CM, Van Herck P, Paelinck B, El Addouli H, La Gerche A, Herbots L, Heidbuchel H, Willems R, Claessen G. Endurance exercise and the risk of cardiovascular pathology in men: a comparison between lifelong and late-onset endurance training and a non-athletic lifestyle - rationale and design of the Master@Heart study, a prospective cohort trial. BMJ Open Sport Exerc Med 7: e001048, 2021. doi:10.1136/bmjsem-2021-001048.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1141. Sung KC, Hong YS, Lee JY, Lee SJ, Chang Y, Ryu S, Zhao D, Cho J, Guallar E, Lima JAC. Physical activity and the progression of coronary artery calcification. Heart 107: 1710–1716, 2021. doi:10.1136/heartjnl-2021-319346.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1142. Zambrano A, Tintut Y, Demer LL, Hsu JJ. Potential mechanisms linking high-volume exercise with coronary artery calcification. Heart 109: 1139–1145, 2023. doi:10.1136/heartjnl-2022-321986.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1143. Pavlovic A, DeFina LF, Leonard D, Radford NB, Farrell SW, Barlow CE, Shuval K, Berry JD, Levine BD. Coronary artery calcification and high-volume physical activity: role of lower intensity vs. longer duration of exercise. Eur J Prev Cardiol 31: 1526–1534, 2024. doi:10.1093/eurjpc/zwae150.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1144. Shuval K, Leonard D, DeFina LF, Barlow CE, Berry JD, Turlington WM, Pavlovic A, Radford NB, Gabriel KP, Khera A, Levine BD. Physical activity and progression of coronary artery calcification in men and women. JAMA Cardiol 9: 659–666, 2024. doi:10.1001/jamacardio.2024.0759.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1145. Papatheodorou E, Aengevaeren VL, Eijsvogels TM, AlFakih K, Hughes RK, Merghani A, Kissel CK, Fyyaz S, Bakalakos A, Wilson MG, Dey D, Finocchiaro G, Parry-Williams G, Torlasco C, Papadakis M, Moon JC, Sharma S. Prevalence of coronary atherosclerosis in female masters endurance athletes. Circulation 150: 1478–1480, 2024. doi:10.1161/CIRCULATIONAHA.124.069484.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1146. Radford NB, DeFina LF, Leonard D, Barlow CE, Willis BL, Gibbons LW, Gilchrist SC, Khera A, Levine BD. Cardiorespiratory fitness, coronary artery calcium, and cardiovascular disease events in a cohort of generally healthy middle-age men: results from the Cooper Center Longitudinal Study. Circulation 137: 1888–1895, 2018. doi:10.1161/CIRCULATIONAHA.117.032708.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1147. Tikkanen E, Gustafsson S, Ingelsson E. Associations of fitness, physical activity, strength, and genetic risk with cardiovascular disease: longitudinal analyses in the UK Biobank Study. Circulation 137: 2583–2591, 2018. doi:10.1161/CIRCULATIONAHA.117.032432.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1148. Feldman DI, Al-Mallah MH, Keteyian SJ, Brawner CA, Feldman T, Blumenthal RS, Blaha MJ. No evidence of an upper threshold for mortality benefit at high levels of cardiorespiratory fitness. J Am Coll Cardiol 65: 629–630, 2015. doi:10.1016/j.jacc.2014.11.030.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1149. Jae SY, Kurl S, Kim HJ, Franklin BA, Kunutsor SK, Kang M, Laukkanen JA. Is there an “asymptote of gain” beyond which further increases in cardiorespiratory fitness convey no additional benefits on mortality and atrial fibrillation? Mayo Clin Proc 94: 545–547, 2019. doi:10.1016/j.mayocp.2018.11.023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1150. Yates T, Razieh C, Henson J, Rowlands AV, Goldney J, Gulsin GS, Davies MJ, Khunti K, Zaccardi F, McCann GP. Device-measured physical activity and cardiac structure by magnetic resonance. Eur Heart J 46: 176–186, 2025. doi:10.1093/eurheartj/ehae506.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1151. Lee DC, Pate RR, Lavie CJ, Sui X, Church TS, Blair SN. Leisure-time running reduces all-cause and cardiovascular mortality risk. J Am Coll Cardiol 64: 472–481, 2014. doi:10.1016/j.jacc.2014.04.058.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1152. Lazarus NR, Harridge SD. Declining performance of master athletes: silhouettes of the trajectory of healthy human ageing? J Physiol 595: 2941–2948, 2017. doi:10.1113/JP272443.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1153. Pedisic Z, Shrestha N, Kovalchik S, Stamatakis E, Liangruenrom N, Grgic J, Titze S, Biddle SJ, Bauman AE, Oja P. Is running associated with a lower risk of all-cause, cardiovascular and cancer mortality, and is the more the better? A systematic review and meta-analysis. Br J Sports Med 54: 898–905, 2020. doi:10.1136/bjsports-2018-100493.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1154. Lee DH, Rezende LF, Joh HK, Keum N, Ferrari G, Rey-Lopez JP, Rimm EB, Tabung FK, Giovannucci EL. Long-term leisure-time physical activity intensity and all-cause and cause-specific mortality: a prospective cohort of US adults. Circulation 146: 523–534, 2022. doi:10.1161/CIRCULATIONAHA.121.058162.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1155. Eijsvogels TM, Molossi S, Lee DC, Emery MS, Thompson PD. Exercise at the extremes: the amount of exercise to reduce cardiovascular events. J Am Coll Cardiol 67: 316–329, 2016. doi:10.1016/j.jacc.2015.11.034.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1156. Levine BD, Baggish AL, Kovacs RJ, Link MS, Maron MS, Mitchell JH; American Heart Association Electrocardiography and Arrhythmias Committee of Council on Clinical Cardiology, Council on Cardiovascular Disease in Young, Council on Cardiovascular and Stroke Nursing, Council on Functional Genomics and Translational Biology, American College of Cardiology. Eligibility and Disqualification Recommendations for Competitive Athletes With Cardiovascular Abnormalities: Task Force 1: Classification of Sports: Dynamic, Static, and Impact: a scientific statement from the American Heart Association and American College of Cardiology. Circulation 132: e262–e266, 2015. doi:10.1161/CIR.0000000000000237.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1157. Foulkes S, Hewitt D, Skow R, Dover D, Kaul P, La Gerche A, Haykowsky M. Outrunning the grim reaper: longevity of the first 200 sub-4 min mile male runners. Br J Sports Med 58: 717–721, 2024. doi:10.1136/bjsports-2024-108386.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1158. Runacres A, Mackintosh KA, McNarry MA. Health Consequences of an elite sporting career: long-term detriment or long-term gain? A meta-analysis of 165,000 former athletes. Sports Med 51: 289–301, 2021. doi:10.1007/s40279-020-01379-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1159. Lemez S, Baker J. Do elite athletes live longer? A systematic review of mortality and longevity in elite athletes. Sports Med Open 1: 16, 2015. doi:10.1186/s40798-015-0024-x.
    Crossref | PubMed | Google Scholar
  • 1160. Garatachea N, Santos-Lozano A, Sanchis-Gomar F, Fiuza-Luces C, Pareja-Galeano H, Emanuele E, Lucia A. Elite athletes live longer than the general population: a meta-analysis. Mayo Clin Proc 89: 1195–1200, 2014. doi:10.1016/j.mayocp.2014.06.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1161. Antero J, Tanaka H, De Larochelambert Q, Pohar-Perme M, Toussaint JF. Female and male US Olympic athletes live 5 years longer than their general population counterparts: a study of 8124 former US Olympians. Br J Sports Med 55: 206–212, 2021. doi:10.1136/bjsports-2019-101696.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1162. Antero-Jacquemin J, Pohar-Perme M, Rey G, Toussaint JF, Latouche A. The heart of the matter: years-saved from cardiovascular and cancer deaths in an elite athlete cohort with over a century of follow-up. Eur J Epidemiol 33: 531–543, 2018. doi:10.1007/s10654-018-0401-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1163. Lin Y, Gajewski A, Poznańska A. Examining mortality risk and rate of ageing among Polish Olympic athletes: a survival follow-up from 1924 to 2012. BMJ Open 6: e010965, 2016. doi:10.1136/bmjopen-2015-010965.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1164. Marijon E, Tafflet M, Antero-Jacquemin J, El Helou N, Berthelot G, Celermajer DS, Bougouin W, Combes N, Hermine O, Empana JP, Rey G, Toussaint JF, Jouven X. Mortality of French participants in the Tour de France (1947–2012). Eur Heart J 34: 3145–3150, 2013. doi:10.1093/eurheartj/eht347.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1165. Kontro TK, Sarna S, Kaprio J, Kujala UM. Mortality and health-related habits in 900 Finnish former elite athletes and their brothers. Br J Sports Med 52: 89–95, 2018. doi:10.1136/bjsports-2017-098206.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1166. Kettunen JA, Kujala UM, Kaprio J, Bäckmand H, Peltonen M, Eriksson JG, Sarna S. All-cause and disease-specific mortality among male, former elite athletes: an average 50-year follow-up. Br J Sports Med 49: 893–897, 2015. doi:10.1136/bjsports-2013-093347.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1167. Clarke PM, Walter SJ, Hayen A, Mallon WJ, Heijmans J, Studdert DM. Survival of the fittest: retrospective cohort study of the longevity of Olympic medallists in the modern era. BMJ 345: e8308, 2012. doi:10.1136/bmj.e8308.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1168. Sanchis-Gomar F, Olaso-Gonzalez G, Corella D, Gomez-Cabrera MC, Vina J. Increased average longevity among the “Tour de France” cyclists. Int J Sports Med 32: 644–647, 2011. doi:10.1055/s-0031-1271711.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1169. Sarna S, Sahi T, Koskenvuo M, Kaprio J. Increased life expectancy of world class male athletes. Med Sci Sports Exerc 25: 237–244, 1993. https://journals.lww.com/acsm-msse/abstract/1993/02000/increased_life_expectancy_of_world_class_male.13.aspx.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1170. Teramoto M, Bungum TJ. Mortality and longevity of elite athletes. J Sci Med Sport 13: 410–416, 2010. doi:10.1016/j.jsams.2009.04.010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1171. Antero-Jacquemin J, Rey G, Marc A, Dor F, Haïda A, Marck A, Berthelot G, Calmat A, Latouche A, Toussaint JF. Mortality in female and male French Olympians: a 1948-2013 cohort study. Am J Sports Med 43: 1505–1512, 2015. doi:10.1177/0363546515574691.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1172. Clarke PM, Walter SJ, Hayen A, Mallon WJ, Heijmans J, Studdert DM. Survival of the fittest: retrospective cohort study of the longevity of Olympic medallists in the modern era. Br J Sports Med 49: 898–902, 2015. doi:10.1136/bjsports-2015-e8308rep.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1173. Booth FW, Roberts CK. Linking performance and chronic disease risk: indices of physical performance are surrogates for health. Br J Sports Med 42: 950–952, 2008. doi:10.1136/bjsm.2008.052589.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1174. Gries KJ, Trappe SW. The aging athlete: paradigm of healthy aging. Int J Sports Med 43: 661–678, 2022. doi:10.1055/a-1761-8481.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1175. Radák Z, Aczél D, Fejes I, Mozaffaritabar S, Pavlik G, Komka Z, Balogh L, Babszki Z, Babszki G, Koltai E, McGreevy KM, Gordevicius J, Horvath S, Kerepesi C. Slowed epigenetic aging in Olympic champions compared to non-champions. Geroscience, 2024. doi:10.1007/s11357-024-01440-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1176. Kujala UM, Marti P, Kaprio J, Hernelahti M, Tikkanen H, Sarna S. Occurrence of chronic disease in former top-level athletes. Predominance of benefits, risks or selection effects? Sports Med 33: 553–561, 2003. doi:10.2165/00007256-200333080-00001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1177. Kujala UM. Is physical activity a cause of longevity? It is not as straightforward as some would believe. A critical analysis. Br J Sports Med 52: 914–918, 2018. doi:10.1136/bjsports-2017-098639.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1178. Bakker EA, Aengevaeren VL, Lee DC, Thompson PD, Eijsvogels TM. All-cause mortality risks among participants in mass-participation sporting events. Br J Sports Med 58: 421–426, 2024. doi:10.1136/bjsports-2023-107190.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1179. Foulkes SJ, Haykowsky MJ, Kistler PM, McConell GK, Trappe S, Hargreaves M, Costill DL, La Gerche A. Lifelong physiology of a former marathon world-record holder: the pros and cons of extreme cardiac remodeling. J Appl Physiol (1985) 137: 461–472, 2024. doi:10.1152/japplphysiol.00070.2024.
    Link | Web of Science | Google Scholar
  • 1180. Kim JH, Malhotra R, Chiampas G, d’Hemecourt P, Troyanos C, Cianca J, Smith RN, Wang TJ, Roberts WO, Thompson PD, Baggish AL; Race Associated Cardiac Arrest Event Registry Study Group. Cardiac arrest during long-distance running races. N Engl J Med 366: 130–140, 2012. doi:10.1056/NEJMoa1106468.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1181. Harris KM, Creswell LL, Haas TS, Thomas T, Tung M, Isaacson E, Garberich RF, Maron BJ. Death and cardiac arrest in U.S. triathlon participants, 1985 to 2016: a case series. Ann Intern Med 167: 529–535, 2017. doi:10.7326/M17-0847.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1182. Mannakkara NN, Finocchiaro G. Exercise and the heart: benefits, risks and adverse effects of exercise training. Rev Cardiovasc Med 24: 94, 2023. doi:10.31083/j.rcm2403094.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1183. Bays-Moneo AB, Izquierdo M, Antón MM, Cadore EL. Cost-consequences analysis following different exercise interventions in institutionalized oldest old: a pilot study of a randomized clinical trial. J Nutr Health Aging 27: 1091–1099, 2023. doi:10.1007/s12603-023-2002-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1184. Morris BA, Sinaei R, Smart NA. Resistance is not futile: a systematic review of the benefits, mechanisms and safety of resistance training in people with heart failure. Heart Fail Rev 29: 827–839, 2024. doi:10.1007/s10741-024-10402-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1185. Baral R, Ho JS, Soroya AN, Hanger M, Clarke RE, Memon SF, Glatzel H, Ahmad M, Providencia R, Bray JJ, D’Ascenzo F. Exercise training improves exercise capacity and quality of life in heart failure with preserved ejection fraction: a systematic review and meta-analysis of randomized controlled trials. Eur Heart J Open 4: oeae033, 2024. doi:10.1093/ehjopen/oeae033.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1186. Franklin BA, Eijsvogels TM. A narrative review on exercise and cardiovascular disease: physical activity thresholds for optimizing health outcomes. Heart Mind 7: 34–39, 2023. doi:10.4103/hm.hm_1_23.
    Crossref | Web of Science | Google Scholar
  • 1187. Lavie CJ, Wisløff U, Blumenthal RS. Extreme physical activity and coronary artery calcification—running heavily and safely with “hearts of stone”. JAMA Cardiol 4: 182–183, 2019. doi:10.1001/jamacardio.2018.4647.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1188. Eijsvogels TMH, Thompson PD, Franklin BA. The “extreme exercise hypothesis”: recent findings and cardiovascular health implications. Curr Treat Options Cardiovasc Med 20: 84, 2018. doi:10.1007/s11936-018-0674-3.
    Crossref | PubMed | Google Scholar
  • 1189. Franklin BA, Thompson PD, Al-Zaiti SS, Albert CM, Hivert MF, Levine BD, Lobelo F, Madan K, Sharrief AZ, Eijsvogels TM; American Heart Association Physical Activity Committee of the Council on Lifestyle and Cardiometabolic Health, Council on Cardiovascular and Stroke Nursing, Council on Clinical Cardiology, Stroke Council. Exercise-Related Acute Cardiovascular Events and Potential Deleterious Adaptations Following Long-Term Exercise Training: Placing the Risks Into Perspective-An Update: a scientific statement from the American Heart Association. Circulation 141: e705–e736, 2020. doi:10.1161/CIR.0000000000000749.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1190. Franklin BA, Kaminsky LA, Kokkinos P. Quantitating the dose of physical activity in secondary prevention: relation of exercise intensity to survival. Mayo Clin Proc 93: 1158–1163, 2018. doi:10.1016/j.mayocp.2018.07.014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1191. Quindry JC, Franklin BA, Chapman M, Humphrey R, Mathis S. Benefits and risks of high-intensity interval training in patients with coronary artery disease. Am J Cardiol 123: 1370–1377, 2019. doi:10.1016/j.amjcard.2019.01.008.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1192. Wewege MA, Ahn D, Yu J, Liou K, Keech A. High-intensity interval training for patients with cardiovascular disease—is it safe? A systematic review. J Am Heart Assoc 7: e009305, 2018. doi:10.1161/JAHA.118.009305.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1193. Franklin BA, Billecke S. Putting the benefits and risks of aerobic exercise in perspective. Curr Sports Med Rep 11: 201–208, 2012. doi:10.1249/JSR.0b013e31825dabd4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1194. Wen CP, Wai JP, Tsai MK, Chen CH. Minimal amount of exercise to prolong life: to walk, to run, or just mix it up? J Am Coll Cardiol 64: 482–484, 2014. doi:10.1016/j.jacc.2014.05.026.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1195. Radaelli R, Rech A, Molinari T, Markarian AM, Petropoulou M, Granacher U, Hortobágyi T, Lopez P. Effects of resistance training volume on physical function, lean body mass and lower-body muscle hypertrophy and strength in older adults: a systematic review and network meta-analysis of 151 randomised trials. Sports Med 55: 167–192, 2025. doi:10.1007/s40279-024-02123-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1196. Glazer NL, Lyass A, Esliger DW, Blease SJ, Freedson PS, Massaro JM, Murabito JM, Vasan RS. Sustained and shorter bouts of physical activity are related to cardiovascular health. Med Sci Sports Exerc 45: 109–115, 2013. doi:10.1249/MSS.0b013e31826beae5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1197. Ross R, Janssen I, Tremblay MS. Public health importance of light intensity physical activity. J Sport Health Sci 13: 674–675, 2024. doi:10.1016/j.jshs.2024.01.010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1198. Wen CP, Wai JP, Tsai MK, Yang YC, Cheng TY, Lee MC, Chan HT, Tsao CK, Tsai SP, Wu X. Minimum amount of physical activity for reduced mortality and extended life expectancy: a prospective cohort study. Lancet 378: 1244–1253, 2011. doi:10.1016/S0140-6736(11)60749-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1199. Lee IM, Powell KE, Sarmiento OL, Hallal PC. Even a small dose of physical activity can be good medicine. Nat Med 31: 376–378, 2025. doi:10.1038/s41591-024-03396-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1200. Hakim AA, Petrovitch H, Burchfiel CM, Ross GW, Rodriguez BL, White LR, Yano K, Curb JD, Abbott RD. Effects of walking on mortality among nonsmoking retired men. N Engl J Med 338: 94–99, 1998. doi:10.1056/NEJM199801083380204.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1201. Kelly P, Williamson C, Niven AG, Hunter R, Mutrie N, Richards J. Walking on sunshine: scoping review of the evidence for walking and mental health. Br J Sports Med 52: 800–806, 2018. doi:10.1136/bjsports-2017-098827.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1202. Murtagh EM, Nichols L, Mohammed MA, Holder R, Nevill AM, Murphy MH. The effect of walking on risk factors for cardiovascular disease: an updated systematic review and meta-analysis of randomised control trials. Prev Med 72: 34–43, 2015. doi:10.1016/j.ypmed.2014.12.041.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1203. Manson JE, Hu FB, Rich-Edwards JW, Colditz GA, Stampfer MJ, Willett WC, Speizer FE, Hennekens CH. A prospective study of walking as compared with vigorous exercise in the prevention of coronary heart disease in women. N Engl J Med 341: 650–658, 1999. doi:10.1056/NEJM199908263410904.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1204. Ahmadi MN, Hamer M, Gill JM, Murphy M, Sanders JP, Doherty A, Stamatakis E. Brief bouts of device-measured intermittent lifestyle physical activity and its association with major adverse cardiovascular events and mortality in people who do not exercise: a prospective cohort study. Lancet Public Health 8: e800–e810, 2023. doi:10.1016/S2468-2667(23)00183-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1205. Bloch-Ibenfeldt M, Theil Gates A, Karlog K, Demnitz N, Kjaer M, Boraxbekk CJ. Heavy resistance training at retirement age induces 4-year lasting beneficial effects in muscle strength: a long-term follow-up of an RCT. BMJ Open Sport Exerc Med 10: e001899, 2024. doi:10.1136/bmjsem-2024-001899.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1206. Jefferis BJ, Parsons TJ, Sartini C, Ash S, Lennon LT, Papacosta O, Morris RW, Wannamethee SG, Lee IM, Whincup PH. Objectively measured physical activity, sedentary behaviour and all-cause mortality in older men: does volume of activity matter more than pattern of accumulation? Br J Sports Med 53: 1013–1020, 2019. doi:10.1136/bjsports-2017-098733.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1207. Millard LAC, Tilling K, Gaunt TR, Carslake D, Lawlor DA. Association of physical activity intensity and bout length with mortality: an observational study of 79,503 UK Biobank participants. PLoS Med 18: e1003757, 2021. doi:10.1371/journal.pmed.1003757.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1208. Saint-Maurice PF, Troiano RP, Matthews CE, Kraus WE. Moderate-to-vigorous physical activity and all-cause mortality: do bouts matter? J Am Heart Assoc 7: e007678, 2018. doi:10.1161/JAHA.117.007678.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1209. Jakicic JM, Kraus WE, Powell KE, Campbell WW, Janz KF, Troiano RP, Sprow K, Torres A, Piercy KL; Physical Activity Guidelines Advisory Committee. Association between bout duration of physical activity and health: systematic review. Med Sci Sports Exerc 51: 1213–1219, 2019. doi:10.1249/MSS.0000000000001933.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1210. Stensvold D, Viken H, Steinshamn SL, Dalen H, Støylen A, Loennechen JP, Reitlo LS, Zisko N, Baekkerud FH, Tari AR, Sandbakk SB, Carlsen T, Ingebrigtsen JE, Lydersen S, Mattsson E, Anderssen SA, Fiatarone Singh MA, Coombes JS, Skogvoll E, Vatten LJ, Helbostad JL, Rognmo O, Wisløff U. Effect of exercise training for five years on all cause mortality in older adults—the Generation 100 study: randomised controlled trial. BMJ 371: m3485, 2020. doi:10.1136/bmj.m3485.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1211. Chen C, Ding S, Wang J. Digital health for aging populations. Nat Med 29: 1623–1630, 2023. doi:10.1038/s41591-023-02391-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1212. Lee IM, Keadle SK, Matthews CE. Fitness trackers to guide advice on activity prescription. JAMA 330: 1733–1734, 2023. doi:10.1001/jama.2023.19332.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1213. Ferguson T, Olds T, Curtis R, Blake H, Crozier AJ, Dankiw K, Dumuid D, Kasai D, O'Connor E, Virgara R, Maher C. Effectiveness of wearable activity trackers to increase physical activity and improve health: a systematic review of systematic reviews and meta-analyses. Lancet Digit Health 4: e615–e626, 2022. doi:10.1016/S2589-7500(22)00111-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1214. Keshet A, Reicher L, Bar N, Segal E. Wearable and digital devices to monitor and treat metabolic diseases. Nat Metab 5: 563–571, 2023. doi:10.1038/s42255-023-00778-y.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1215. Khurshid S, Weng LC, Nauffal V, Pirruccello JP, Venn RA, Al-Alusi MA, Benjamin EJ, Ellinor PT, Lubitz SA. Wearable accelerometer-derived physical activity and incident disease. NPJ Digit Med 5: 131, 2022. doi:10.1038/s41746-022-00676-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1216. Slemenšek J, Fister I, Geršak J, Bratina B, van Midden VM, Pirtošek Z, Šafarič R. Human gait activity recognition machine learning methods. Sensors (Basel) 23: 745, 2023. doi:10.3390/s23020745.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1217. Yu S, Chen Z, Wu X. The impact of wearable devices on physical activity for chronic disease patients: findings from the 2019 Health Information National Trends Survey. Int J Environ Res Public Health 20: 887, 2023. doi:10.3390/ijerph20010887.
    Crossref | Web of Science | Google Scholar
  • 1218. Girolami M, Mavilia F, Delmastro F. Sensing social interactions through BLE beacons and commercial mobile devices. Pervasive Mob Comput 67: 101198, 2020. doi:10.1016/j.pmcj.2020.101198.
    Crossref | PubMed | Google Scholar
  • 1219. Shei RJ, Holder IG, Oumsang AS, Paris BA, Paris HL. Wearable activity trackers—advanced technology or advanced marketing? Eur J Appl Physiol 122: 1975–1990, 2022. doi:10.1007/s00421-022-04951-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1220. Seneviratne MG, Connolly SB, Martin SS, Parakh K. Grains of sand to clinical pearls: realizing the potential of wearable data. Am J Med 136: 136–142, 2023. doi:10.1016/j.amjmed.2022.10.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1221. Lima FV, Kadiyala V, Huang A, Agusala K, Cho D, Freeman AM, Druz R. At the crossroads! Time to start taking smartwatches seriously. Am J Cardiol 179: 96–101, 2022. doi:10.1016/j.amjcard.2022.06.020.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1222. Kang HS, Exworthy M. Wearing the future—wearables to empower users to take greater responsibility for their health and care: scoping review. JMIR Mhealth Uhealth 10: e35684, 2022. doi:10.2196/35684.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1223. Santana A, Mediano M, Kasal D. Physical performance tests and in-hospital outcomes in elective open chest heart surgery. Int J Cardiol Heart Vasc 44: 101164, 2023. doi:10.1016/j.ijcha.2022.101164.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1224. Staes M, Gyselinck I, Goetschalckx K, Troosters T, Janssens W. Identifying limitations to exercise with incremental cardiopulmonary exercise testing: a scoping review. Eur Respir Rev 33: 240010, 2024. doi:10.1183/16000617.0010-2024.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1225. Brawner CA, Ehrman JK, Keteyian SJ. Are international standards for exercise capacity ready for prime time? Mayo Clin Proc 95: 218–220, 2020. doi:10.1016/j.mayocp.2019.12.018.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1226. Andonian BJ, Hardy N, Bendelac A, Polys N, Kraus WE. Making cardiopulmonary exercise testing interpretable for clinicians. Curr Sports Med Rep 20: 545–552, 2021. doi:10.1249/JSR.0000000000000895.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1227. Sidhu SS, Saggar K, Goyal O, Kishore H, Sidhu SS. Normative values of skeletal muscle mass, strength and performance in the Indian population. Indian J Gastroenterol 43: 628–637, 2024. doi:10.1007/s12664-024-01565-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1228. Roman-Liu D, Kamińska J, Tokarski TM. Population-specific equations of age-related maximum handgrip force: a comprehensive review. PeerJ 12: e17703, 2024. doi:10.7717/peerj.17703.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1229. Swift DL, Johannsen NM, Earnest CP, Newton RL Jr, McGee JE, Church TS. Cardiorespiratory fitness and exercise training in African Americans. Prog Cardiovasc Dis 60: 96–102, 2017. doi:10.1016/j.pcad.2017.06.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1230. Swift DL, Staiano AE, Johannsen NM, Lavie CJ, Earnest CP, Katzmarzyk PT, Blair SN, Newton RL Jr, Church TS. Low cardiorespiratory fitness in African Americans: a health disparity risk factor? Sports Med 43: 1301–1313, 2013. doi:10.1007/s40279-013-0092-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1231. O CK, Chan JC. Insights into optimal BMI from the GlasVEGAS study. Nat Metab 6: 1435–1437, 2024. doi:10.1038/s42255-024-01088-7.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1232. Koch B, Schäper C, Ittermann T, Spielhagen T, Dörr M, Völzke H, Opitz CF, Ewert R, Gläser S. Reference values for cardiopulmonary exercise testing in healthy volunteers: the SHIP study. Eur Respir J 33: 389–397, 2009. doi:10.1183/09031936.00074208.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1233. Chen LK, Meng LC, Peng LN, Lee WJ, Zhang S, Nishita Y, Otsuka R, Yamada M, Pan WH, Kamaruzzaman SB, Woo J, Hsiao FY, Arai H. Mapping normative muscle health metrics across the aging continuum: a multinational study pooling data from eight cohorts in Japan, Malaysia and Taiwan. J Cachexia Sarcopenia Muscle 16: e13731, 2025. doi:10.1002/jcsm.13731.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1234. Kaminsky LA, Arena R, Beckie TM, Brubaker PH, Church TS, Forman DE, Franklin BA, Gulati M, Lavie CJ, Myers J, Patel MJ, Pina IL, Weintraub WS, Williams MA; American Heart Association Advocacy Coordinating Committee, Council on Clinical Cardiology, Council on Nutrition, Physical Activity and Metabolism. The importance of cardiorespiratory fitness in the United States: the need for a national registry: a policy statement from the American Heart Association. Circulation 127: 652–662, 2013. doi:10.1161/CIR.0b013e31827ee100.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1235. Kaminsky LA, Myers J, Brubaker PH, Franklin BA, Bonikowske AR, German C, Arena R. 2023 update: The importance of cardiorespiratory fitness in the United States. Prog Cardiovasc Dis 83: 3–9, 2024. doi:10.1016/j.pcad.2024.01.020.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1236. Western MJ, Malkowski OS. Associations of the Short Physical Performance Battery (SPPB) with adverse health outcomes in older adults: a 14-year follow-up from the English Longitudinal Study of Ageing (ELSA). Int J Environ Res Public Health 19: 16319, 2022. doi:10.3390/ijerph192316319.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1237. Furrer R, van Schoor NM, de Haan A, Lips P, de Jongh RT. Gender-specific associations between physical functioning, bone quality, and fracture risk in older people. Calcif Tissue Int 94: 522–530, 2014. doi:10.1007/s00223-013-9836-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1238. Gichu M, Harwood RH. Measurement of healthy ageing. Age Ageing 52: iv3–iv5, 2023. doi:10.1093/ageing/afad118.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1239. Tavassoli N, de Souto Barreto P, Berbon C, Mathieu C, de Kerimel J, Lafont C, Takeda C, Carrie I, Piau A, Jouffrey T, Andrieu S, Nourhashemi F, Beard JR, Soto Martin ME, Vellas B. Implementation of the WHO integrated care for older people (ICOPE) programme in clinical practice: a prospective study. Lancet Healthy Longev 3: e394–e404, 2022. doi:10.1016/S2666-7568(22)00097-6.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1240. Beard JR, Jotheeswaran AT, Cesari M, Araujo de Carvalho I. The structure and predictive value of intrinsic capacity in a longitudinal study of ageing. BMJ Open 9: e026119, 2019. doi:10.1136/bmjopen-2018-026119.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1241. Thiyagarajan JA, Araujo de Carvalho I, Peña-Rosas JP, Chadha S, Mariotti SP, Dua T, Albanese E, Bruyère O, Cesari M, Dangour A, Dias A, Guerra M, Keeffe J, Kerse N, Khan QU, Liu CJ, Murthy GV, Ndegwa SN, Reginster JY, Robledo L, Tremblay K, Woo J, Prince M, Beard JR. Redesigning care for older people to preserve physical and mental capacity: WHO guidelines on community-level interventions in integrated care. PLoS Med 16: e1002948, 2019. doi:10.1371/journal.pmed.1002948.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1242. Flammer J, Konieczka K, Bruno RM, Virdis A, Flammer AJ, Taddei S. The eye and the heart. Eur Heart J 34: 1270–1278, 2013. doi:10.1093/eurheartj/eht023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1243. Nusinovici S, Rim TH, Li H, Yu M, Deshmukh M, Quek TC, Lee G, Chong CCY, Peng Q, Xue CC, Zhu Z, Chew EY, Sabanayagam C, Wong TY, Tham YC, Cheng CY. Application of a deep-learning marker for morbidity and mortality prediction derived from retinal photographs: a cohort development and validation study. Lancet Healthy Longev 5: 100593, 2024. doi:10.1016/S2666-7568(24)00089-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1244. Yusufu M, Friedman DS, Kang M, Padhye A, Shang X, Zhang L, Shi D, He M. Retinal vascular fingerprints predict incident stroke: findings from the UK Biobank cohort study. Heart 111: 306–313, 2025. doi:10.1136/heartjnl-2024-324705.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1245. Chaput JP, Biswas RK, Ahmadi M, Cistulli PA, Rajaratnam SM, Bian W, St-Onge MP, Stamatakis E. Sleep regularity and major adverse cardiovascular events: a device-based prospective study in 72 269 UK adults. J Epidemiol Community Health 79: 257–264, 2025. doi:10.1136/jech-2024-222795.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1246. Ulfa M, Setyonugroho W, Lestari T, Widiasih E, Nguyen Quoc A. Nutrition-related mobile application for daily dietary self-monitoring. J Nutr Metab 2022: 2476367, 2022. doi:10.1155/2022/2476367.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1247. Niemelä M, Maijala A, Nauha L, Jämsä T, Korpelainen R, Farrahi V. Associations of wearable ring measured sleep, sedentary time, and physical activity with cardiometabolic health: a compositional data analysis approach. Scand J Med Sci Sports 34: e14710, 2024. doi:10.1111/sms.14710.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1248. Bajunaid R, Niu C, Hambly C, Liu Z, Yamada Y, Aleman-Mateo H, , et al.. Predictive equation derived from 6,497 doubly labelled water measurements enables the detection of erroneous self-reported energy intake. Nat Food 6: 58–71, 2025. doi:10.1038/s43016-024-01089-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1249. Isakadze N, Martin SS. How useful is the smartwatch ECG? Trends Cardiovasc Med 30: 442–448, 2020. doi:10.1016/j.tcm.2019.10.010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1250. Keshet A, Shilo S, Godneva A, Talmor-Barkan Y, Aviv Y, Segal E, Rossman H. CGMap: characterizing continuous glucose monitor data in thousands of non-diabetic individuals. Cell Metab 35: 758–769.e3, 2023. doi:10.1016/j.cmet.2023.04.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1251. Park H, Metwally AA, Delfarah A, Wu Y, Perelman D, Rodgar M, Mayer C, Celli A, McLaughlin T, Mignot E, Snyder M. Lifestyle profiling using wearables and prediction of glucose metabolism in individuals with normoglycemia or prediabetes (Preprint). medRxiv 2024.09.05.24312545, 2024. doi:10.1101/2024.09.05.24312545.
    Google Scholar
  • 1252. Huang N, Ge M, Liu X, Tian X, Yin P, Bao Z, Cao F, Shyh-Chang N, Dong B, Dai L, Gan Z, Hu P, Qu J, Wang S, Wang H, Xiao Q, Yue R, Yue J, Zhang L, Zhang Y, Zhang H, Zhang W, Liu GH, Pei G, Liu Y, Zhu D, Dong B; Aging Biomarker Consortium. A framework of biomarkers for skeletal muscle aging: a consensus statement by the Aging Biomarker Consortium. Life Med 3: lnaf001, 2024. doi:10.1093/lifemedi/lnaf001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1253. Ortega FB, Cadenas-Sanchez C, Lee DC, Ruiz JR, Blair SN, Sui X. Fitness and fatness as health markers through the lifespan: an overview of current knowledge. Prog Prev Med (NY) 3: e0013, 2018. doi:10.1097/pp9.0000000000000013.
    Crossref | PubMed | Google Scholar
  • 1254. Zhao W, Dai C, Wang Q, Zhang J, Lou X, Chen R, Shen G, Zhang Y. Sarcopenia risk in U.S. younger adults: the impact of physical activity intensity and occupational engagement-insights from a cross-sectional NHANES study. BMC Public Health 24: 3179, 2024. doi:10.1186/s12889-024-20665-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1255. Evans WJ, Guralnik J, Cawthon P, Appleby J, Landi F, Clarke L, Vellas B, Ferrucci L, Roubenoff R. Sarcopenia: no consensus, no diagnostic criteria, and no approved indication—how did we get here? Geroscience 46: 183–190, 2024. doi:10.1007/s11357-023-01016-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1256. Chen LK. Community-powered actions building a healthier future for aging populations. Arch Gerontol Geriatr 128: 105652, 2025. doi:10.1016/j.archger.2024.105652.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1257. Diaz T, Banerjee A. Welcome to the supplement on measurement of healthy ageing. Age Ageing 52: iv1–iv2, 2023. doi:10.1093/ageing/afad103.
    Crossref | Web of Science | Google Scholar
  • 1258. Vazquez-Guajardo M, Rivas D, Duque G. Exercise as a therapeutic tool in age-related frailty and cardiovascular disease: challenges and strategies. Can J Cardiol 40: 1458–1467, 2024. doi:10.1016/j.cjca.2024.01.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1259. Hills AP, Jayasinghe S, Arena R, Byrne NM. Global status of cardiorespiratory fitness and physical activity—are we improving or getting worse? Prog Cardiovasc Dis 83: 16–22, 2024. doi:10.1016/j.pcad.2024.02.008.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1260. Wang Z, Emmerich A, Pillon NJ, Moore T, Hemerich D, Cornelis MC, , et al.. Genome-wide association analyses of physical activity and sedentary behavior provide insights into underlying mechanisms and roles in disease prevention. Nat Genet 54: 1332–1344, 2022. doi:10.1038/s41588-022-01165-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1261. Ramirez FD, Chen Y, Di Santo P, Simard T, Motazedian P, Hibbert B. Association between self-reported potentially modifiable cardiac risk factors and perceived need to improve physical health: a population-based study. J Am Heart Assoc 6: e005491, 2017. doi:10.1161/JAHA.117.005491.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1262. Garcia-Hermoso A, López-Gil JF, Ramírez-Vélez R, Alonso-Martínez AM, Izquierdo M, Ezzatvar Y. Adherence to aerobic and muscle-strengthening activities guidelines: a systematic review and meta-analysis of 3.3 million participants across 32 countries. Br J Sports Med 57: 225–229, 2023. doi:10.1136/bjsports-2022-106189.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1263. Lehtonen E, Gagnon D, Eklund D, Kaseva K, Peltonen JE. Hierarchical framework to improve individualised exercise prescription in adults: a critical review. BMJ Open Sport Exerc Med 8: e001339, 2022. doi:10.1136/bmjsem-2022-001339.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1264. Morgan PJ, Young MD, Smith JJ, Lubans DR. Targeted health behavior interventions promoting physical activity: a conceptual model. Exerc Sport Sci Rev 44: 71–80, 2016. doi:10.1249/JES.0000000000000075.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1265. Pratt M, Ramirez Varela A, Salvo D, Kohl Iii HW, Ding D. Attacking the pandemic of physical inactivity: what is holding us back? Br J Sports Med 54: 760–762, 2020. doi:10.1136/bjsports-2019-101392.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1266. Collins KA, Huffman KM, Wolever RQ, Smith PJ, Siegler IC, Ross LM, Hauser ER, Jiang R, Jakicic JM, Costa PT, Kraus WE. Determinants of dropout from and variation in adherence to an exercise intervention: the STRRIDE randomized trials. Transl J Am Coll Sports Med 7: e000190, 2022. doi:10.1249/tjx.0000000000000190.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1267. Zhu S, Sinha D, Kirk M, Michalopoulou M, Hajizadeh A, Wren G, Doody P, Mackillop L, Smith R, Jebb SA, Astbury NM. Effectiveness of behavioural interventions with motivational interviewing on physical activity outcomes in adults: systematic review and meta-analysis. BMJ 386: e078713, 2024. doi:10.1136/bmj-2023-078713.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1268. Ness RG, Patton RW. The effect of beliefs on maximum weight-lifting performance. Cogn Ther Res 3: 205–211, 1979. doi:10.1007/BF01172606.
    Crossref | Web of Science | Google Scholar
  • 1269. Feil K, Fritsch J, Rhodes RE. The intention-behaviour gap in physical activity: a systematic review and meta-analysis of the action control framework. Br J Sports Med 57: 1265–1271, 2023. doi:10.1136/bjsports-2022-106640.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1270. Rhodes RE. Translating physical activity intentions into behavior: reflective, regulatory, and reflexive processes. Exerc Sport Sci Rev 52: 13–22, 2024. doi:10.1249/JES.0000000000000329.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1271. WHO. Global action plan on physical activity 2018–2030: more active people for a healthier world. World Health Organization, 2018. https://www.who.int/publications/i/item/9789241514187.
    Google Scholar
  • 1272. Sherwood NE, Jeffery RW. The behavioral determinants of exercise: implications for physical activity interventions. Annu Rev Nutr 20: 21–44, 2000. doi:10.1146/annurev.nutr.20.1.21.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1273. Westerbeek H, Eime R. The Physical Activity and Sport Participation Framework—a policy model toward being physically active across the lifespan. Front Sports Act Living 3: 608593, 2021. doi:10.3389/fspor.2021.608593.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1274. Rutters F, den Braver NR, Lakerveld J, Mackenbach JD, van der Ploeg HP, Griffin S, Elders PJM, Beulens JW. Lifestyle interventions for cardiometabolic health. Nat Med 30: 3455–3467, 2024. doi:10.1038/s41591-024-03373-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1275. Bista S, Debache I, Chaix B. Physical activity and sedentary behaviour related to transport activity assessed with multiple body-worn accelerometers: the RECORD MultiSensor Study. Public Health 189: 144–152, 2020. doi:10.1016/j.puhe.2020.10.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1276. Chakrabarti S, Shin EJ. Automobile dependence and physical inactivity: insights from the California Household Travel Survey. J Transp Health 6: 262–271, 2017. doi:10.1016/j.jth.2017.05.002.
    Crossref | Web of Science | Google Scholar
  • 1277. Sugiyama T, Chandrabose M, Homer AR, Sugiyama M, Dunstan DW, Owen N. Car use and cardiovascular disease risk: Systematic review and implications for transport research. J Transp Health 19: 100930, 2020. doi:10.1016/j.jth.2020.100930.
    Crossref | Web of Science | Google Scholar
  • 1278. Mackay A, Mackay DF, Celis-Morales CA, Lyall DM, Gray SR, Sattar N, Gill JM, Pell JP, Anderson JJ. The association between driving time and unhealthy lifestyles: a cross-sectional, general population study of 386 493 UK Biobank participants. J Public Health (Oxf) 41: 527–534, 2019. doi:10.1093/pubmed/fdy155.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1279. Wang ML, Narcisse MR, McElfish PA. Higher walkability associated with increased physical activity and reduced obesity among United States adults. Obesity (Silver Spring) 31: 553–564, 2023. doi:10.1002/oby.23634.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1280. Narcisse MR, Wang ML, Schootman M, DelNero P, Schwarz AG, McElfish PA. Physical activity among cancer survivors: do neighborhood walkability and metropolitan size play a role? J Cancer Surviv, 2024. doi:10.1007/s11764-024-01584-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1281. Collado-Mateo D, Lavín-Pérez AM, Peñacoba C, Del Coso J, Leyton-Román M, Luque-Casado A, Gasque P, Fernández-Del-Olmo MA, Amado-Alonso D. Key factors associated with adherence to physical exercise in patients with chronic diseases and older adults: an umbrella review. Int J Environ Res Public Health 18: 2023, 2021. doi:10.3390/ijerph18042023.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1282. Teixeira DS, Bastos V, Andrade AJ, Palmeira AL, Ekkekakis P. Individualized pleasure-oriented exercise sessions, exercise frequency, and affective outcomes: a pragmatic randomized controlled trial. Int J Behav Nutr Phys Act 21: 85, 2024. doi:10.1186/s12966-024-01636-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1283. Lee AK, Muhamad RB, Tan VP. Physically active primary care physicians consult more on physical activity and exercise for patients: a public teaching-hospital study. Sports Med Health Sci 6: 82–88, 2024. doi:10.1016/j.smhs.2023.11.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1284. Izquierdo M, Duque G, Morley JE. Physical activity guidelines for older people: knowledge gaps and future directions. Lancet Healthy Longev 2: e380–e383, 2021. doi:10.1016/S2666-7568(21)00079-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1285. Izquierdo M, Merchant RA, Morley JE, Anker SD, Aprahamian I, Arai H, , et al.. International Exercise Recommendations in Older Adults (ICFSR): expert consensus guidelines. J Nutr Health Aging 25: 824–853, 2021. doi:10.1007/s12603-021-1665-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1286. Palumbo M, Modena R, Bortolan L, Skafidas S, Callovini A, Savoldelli A, Gilli F, Fornasiero A, Schena F, Pellegrini B, Zoppirolli C. Effects of a similar amount of regular non-structured or competitive physical activity across late adulthood: a cross-sectional study. Front Sports Act Living 6: 1416080, 2024. doi:10.3389/fspor.2024.1416080.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1287. Tiller NB, Stellingwerff T, Witard OC, Hawley JA, Burke LM, Betts JA. The nontechnical summary: a new initiative to enhance the translation of sports science research and reduce the spread of misinformation. Int J Sport Nutr Exerc Metab 34: 337–339, 2024. doi:10.1123/ijsnem.2024-0151.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1288. Sadana R, Blas E, Budhwani S, Koller T, Paraje G. Healthy ageing: raising awareness of inequalities, determinants, and what could be done to improve health equity. Gerontologist 56, Suppl 2: S178–S193, 2016. doi:10.1093/geront/gnw034.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1289. Ding D, Chastin S, Salvo D, Nau T, Gebel K, Sanchez-Lastra MA, Luo M, Crochemore-Silva I, Ekelund U, Bauman A. Realigning the physical activity research agenda for population health, equity, and wellbeing. Lancet 404: 411–414, 2024. doi:10.1016/S0140-6736(24)01540-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1290. Wackerhage H, Schoenfeld BJ. Personalized, evidence-informed training plans and exercise prescriptions for performance, fitness and health. Sports Med 51: 1805–1813, 2021. doi:10.1007/s40279-021-01495-w.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1291. Grindem H, Myklebust G. Be a champion for your athlete’s health. J Orthop Sports Phys Ther 50: 173–175, 2020. doi:10.2519/jospt.2020.0605.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1292. Guess N. Big data and personalized nutrition: the key evidence gaps. Nat Metab 6: 1420–1422, 2024. doi:10.1038/s42255-023-00960-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1293. Ding D, Lawson KD, Kolbe-Alexander TL, Finkelstein EA, Katzmarzyk PT, van Mechelen W, Pratt M; Lancet Physical Activity Series 2 Executive Committee. The economic burden of physical inactivity: a global analysis of major non-communicable diseases. Lancet 388: 1311–1324, 2016. doi:10.1016/S0140-6736(16)30383-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1294. Santos AC, Willumsen J, Meheus F, Ilbawi A, Bull FC. The cost of inaction on physical inactivity to public health-care systems: a population-attributable fraction analysis. Lancet Glob Health 11: e32–e39, 2023. doi:10.1016/S2214-109X(22)00464-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1295. Wang Y, Müller J, Myers J. Association between cardiorespiratory fitness and health care costs in hypertensive men. Atherosclerosis 331: 1–5, 2021. doi:10.1016/j.atherosclerosis.2021.06.914.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1296. Myers J, de Souza ES, Doom R, Fonda H, Chan K, Kamil-Rosenberg S, Kokkinos P. Cardiorespiratory fitness and health care costs in diabetes: the Veterans Exercise Testing Study. Am J Med 132: 1084–1090, 2019. doi:10.1016/j.amjmed.2019.04.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1297. Morera A, Calatayud J, López-Bueno R, Núñez-Cortés R, Bláfoss R, Venge Skovlund S, Andersen LL. Leisure-time physical activity to reduce risk of long-term sickness absence across diverse subgroups in the working population-a prospective cohort study of 68,000 participants. J Phys Act Health 22: 85–91, 2025. doi:10.1123/jpah.2024-0352.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1298. Scrafford CG, Bi X, Multani JK, Murphy MM, Schmier JK, Barraj LM. Health economic evaluation modeling shows potential health care cost savings with increased conformance with healthy dietary patterns among adults in the United States. J Acad Nutr Diet 119: 599–616, 2019. doi:10.1016/j.jand.2018.10.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1299. Jackson T. The False Economy of Big Food— and the case for a new food economy. The Food, Farming and Countryside Commission (FFCC). https://ffcc.co.uk/publications/the-false-economy-of-big-food [2024 Nov 22].
    Google Scholar
  • 1300. Gössling S, Choi A, Dekker K, Metzler D. The social cost of automobility, cycling and walking in the European Union. Ecol Econ 158: 65–74, 2019. doi:10.1016/j.ecolecon.2018.12.016.
    Crossref | Web of Science | Google Scholar
  • 1301. Chaput JP, Carrier J, Bastien C, Gariépy G, Janssen I. Economic burden of insufficient sleep duration in Canadian adults. Sleep Health 8: 298–302, 2022. doi:10.1016/j.sleh.2022.02.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1302. Chaput JP, Janssen I, Sampasa-Kanyinga H, Tomkinson GR, Lang JJ. Economic burden of low cardiorespiratory fitness in Canada. Prev Med 168: 107424, 2023. doi:10.1016/j.ypmed.2023.107424.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1303. Chaput JP, Janssen I, Sampasa-Kanyinga H, Tomkinson GR, Lang JJ. Economic burden of low muscle strength in Canadian adults. Appl Physiol Nutr Metab 48: 634–638, 2023. doi:10.1139/apnm-2022-0371.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1304. Burns ER, Stevens JA, Lee R. The direct costs of fatal and non-fatal falls among older adults—United States. J Safety Res 58: 99–103, 2016. doi:10.1016/j.jsr.2016.05.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1305. Lee J, Jukarainen S, Karvanen A, Dixon P, Davies NM, Smith GD, Natarajan P, Ganna A. Quantifying the causal impact of biological risk factors on healthcare costs. Nat Commun 14: 5672, 2023. doi:10.1038/s41467-023-41394-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1306. Elliehausen CJ, Anderson RM, Diffee GM, Rhoads TW, Lamming DW, Hornberger TA, Konopka AR. Geroprotector drugs and exercise: friends or foes on healthy longevity? BMC Biol 21: 287, 2023. doi:10.1186/s12915-023-01779-9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1307. Furrer R, Handschin C. Biomarkers of aging: functional aspects still trump molecular parameters. NPJ Aging 11: 15, 2025. doi:10.1038/s41514-025-00207-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1308. Johnson BT, Acabchuk RL. What are the keys to a longer, happier life? Answers from five decades of health psychology research. Soc Sci Med 196: 218–226, 2018. doi:10.1016/j.socscimed.2017.11.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1309. Perri G, French C, Agostinis-Sobrinho C, Anand A, Antarianto RD, Arai Y, , et al.. An expert consensus statement on biomarkers of ageing for use in intervention studies. J Gerontol A Biol Sci Med Sci 2024: glae297, 2024. doi:10.1093/gerona/glae297.
    Crossref | Web of Science | Google Scholar
  • 1310. Scott AJ, Ellison M, Sinclair DA. The economic value of targeting aging. Nat Aging 1: 616–623, 2021. doi:10.1038/s43587-021-00080-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 1311. Gems D, Okholm S, Lemoine M. Inflated expectations: the strange craze for translational research on aging: given existing confusion about the basic science of aging, why the high optimism in the private sector about the prospects of developing anti-aging treatments? EMBO Rep 25: 3748–3752, 2024. doi:10.1038/s44319-024-00226-2.
    Crossref | PubMed | Web of Science | Google Scholar

AUTHOR NOTES