What is meant by biological aging?

Recognize which are the elements that predict why a person is aging faster or slower and which intervention we can arrange to slow down the process, which permits to prevent or delay the progression of multimorbidity and disability.

Recent Findings

Aging is a complex process that leads to changes in all the systems of the body and all the functions of the person; however, aging develops at different rates in different people, and chronological age is not always consistent with biological age.

Summary

Gerontologists are focused not only on finding the best theory able to explain aging but also on identifying one or more markers, which are able to describe aging processes. These biomarkers are necessary to better define the aging-related pathologies, manage multimorbidity, and improve the quality of life. The aim of this paper is to review the most recent evidence on aging biomarkers and the clusters related to them for personalization of treatments.

Working on a manuscript?

Avoid the common mistakes

Introduction

“Most people don’t grow up. Most people age. They find parking spaces, honour their credit cards, get married, have children, and call that maturity. What that is, is aging.”—Maya Angelou. One of the biggest megatrends impacting the world today is population aging. Aging is a topic that has captivated both scientists and philosophers throughout history, but aging as a population scenario emerged on a worldwide scale for the first time in the last century. Thus, it is hard to really identify a definition of aging. It is a decrease in fitness with chronological age, it is a developmental phase beyond the normal life trajectory and it is a time of the increased risk of physical and psychological disabilities testing the limits of resilience.

Aging occurs at a different rate in varying geographic regions of the world.

Europe is currently the oldest region, with 17.4% of the total population aged 65 and older. However, the Asia and Latin America older population is growing fast, with Asia’s older population almost tripling in size from 341.4 million in 2015 to 975.3 million in 2050 [].

All these data do not consider aging as an epiphenomenon, but an individual data of the global population, just a chronological number. Aging is intrinsically a complex scenario characterized by changes that take place at different levels of biological systems. Biological age is of course influenced by chronological age, but chronological age is by itself not representative of biological age; biological age is determined by physiological reserve and functional status. Assessing biological age is essential to predict life expectancy and resilience to stressors []. If any definition of aging may appear incomplete and insufficient, much more difficult and complex is to find the marker (or biomarker) that can identify it.

Many theories currently trying to explain aging processes and many biomarkers are identified to measure aging and its evolutionary stages. Theories and biomarkers are not studied to extend life span but to guide therapeutic choices and optimize patient management and personalization of care.

The purpose of this paper is not purely to list which biomarkers are able to identify the various stages of aging, rather explain how an epiphenomenon, natural and physiological, is so complex [], how many factors are protagonists in its development, and how many actors and characters play in maximizing its individual features, taking into account social and morbidity biomarker. These factors, such as frailty, loss of autonomy, essential needs, and comorbidities, influence the aging process and are able to justify why the biological age of a person living in a country does not correspond to the age of another person living in a country with better socio-sanitary conditions.

Clinical and Biological Aging Phenotypes

The aging phenotype can be described as a complex mosaic resulting from the interaction of a variety of environmental, stochastic, and genetic–epigenetic events/stimuli impinging lifelong on our body [, ].

There is no clear evidence which molecular, cellular, or physiological changes are the most important drivers of the aging process and/or how they influence one another []. In its broadest sense, aging merely refers to the changes that occur during an organisms’ life span, though the rate at which these take place varies widely []. Despite its enormous complexity, involving combinations of these variables, a small number of basic molecular mechanisms underpin the aging process, including a set of evolutionary highly conserved basic biological mechanisms responsible for body maintenance and repair. One of the key mechanisms is inflammation; a typical feature of the aging process is the development of a chronic, low-grade inflammatory status named “inflammaging” [•], which emerged as critical in the pathogenesis of major age-related chronic diseases such as atherosclerosis, type 2 diabetes, and neurodegeneration. Inflammaging plays a pivotal role in the most important geriatric conditions, such as sarcopenia [••], osteoporosis [], frailty, and disability, thus contributing to mortality []. Interestingly, a variety of tissues (adipose tissue, muscle), organs (brain, liver), systems (immune system), and ecosystems (gut microbiota) of the body (indicated as “sub-systems”) can contribute to the onset and progression of such a systemic inflammatory state [] by increasing the production of several pro-inflammatory mediators or lowering that of the anti-inflammatory ones [•].

To differentiate the innocuous changes from those leading to increased risk of disease, disability, or death, bio-gerontologists tend to use a more precise term—senescence—when describing aging []. Senescence is, therefore, the progressive deterioration of bodily functions over time and normal human aging has been associated with a loss of complexity in a wide range of physiological processes and anatomic structures [], including blood pressure [], stride intervals [], respiratory cycles [], and vision [], among others, such as postural dynamics [], ultimately leading to decreased fertility and increased risk or mortality [].

Systemic consequences of aging are widespread but they can be clustered into four domains (Fig. ):

  • Changing in body composition

  • The balance between energy availability and energy demand

  • Signaling networks that maintain homeostasis

  • Neurodegeneration

Fig. 1

What is meant by biological aging?

Systemic consequences of aging

Full size image

These changes develop in parallel and affect each other through many feed-forward and feedback loop.

The phenotype that results from the aging process is characterized by increased susceptibility to disease, high risk of multiple coexisting diseases, impaired response to stress, the emergence of “geriatric syndromes,” altered response to treatment, high risk of disability, and loss of personal autonomy with all its psychological and social consequences. On the other hand, all these factors influence aging itself, in a dynamic and parallel way, so that they can be considered as not only a consequence of aging but also an integral part of the aging process.

Theories of Aging

Human aging is currently defined as a dynamic process involving the continual adaptation of the body to lifelong exposure to internal and external damaging, as conceptualized in the “remodelling theory of aging” [••]. Theories of aging are generally classified as either program or damage theories. Programmed aging theories suggest that there is a deliberate deterioration with age because a limited life span results in evolutionary benefits []. This plan could be a result of “aging genes.” The first described mutation to yield a significant extension in the life span of Caenorhabditis elegans was in the age-I gene, which was shown to result in a 65% increase in mean life span and a 110% increase in maximum life span of this organism []. Evolutionary biologists may argue that aging occurs due to the absence of natural selection at the post-reproductive stage of life []. Although such aging theories are subjectively appealing, as they convey a cure for aging, the accumulation of damage is a spontaneous entropy-driven process []. Among the damage theories, a prevailing idea is that of oxidative damage. Reactive oxygen species (ROS) are generated during metabolism through several interrelated reactions. The supposition that aging may be caused by ROS has been further substantiated by studies involving transgenic animals for genes encoding antioxidants. The life span of Drosophila melanogaster has been extended by overexpression of both superoxide dismutase (SOD) and catalase, both antioxidant enzymes []. Since mitochondria are the major producer of ROS in mammalian cells, mitochondrial DNA (mtDNA) is therefore particularly susceptible to oxidative damage []. Mitochondrial maintenance is, therefore, essential to preserve cellular homeostasis and impaired mitochondrial maintenance has been described as a shared hallmark of numerous human pathologies and aging []. Mitochondrial DNA varies with age, and it is commonly considered that DNA hypomethylation is a typical aspect of the aging process []. ROS are active intermediates of DNA methylation, as well as histone modification. These reactive oxygen species may play a role in epigenetic processes (physiological phenotypic variations caused by external or environmental factors that switch genes on/off) through reactions of nucleophilic substitution at the DNA level. Consequently, it has been suggested that better preservation of DNA methylation levels, slower cell metabolism, and improved control in signal transmission through epigenetic mechanisms could be key processes involved in human longevity. Oxidative damage to proteins is irreversible and irreparable [] and must be degraded by the proteasome. The proteasome is the most important proteolytic machinery in eukaryotic cells, largely responsible for the removal of oxidized proteins and the prevention of its aggregation []. However, it has been shown that the activity of proteasome is impaired during aging leading to the accumulation of oxidizing proteins, aggresome and lipofuscin, so-called the age pigment. Similarly, to oxidative damage, nitrosamine damage—that caused by reactive nitrogen species (RNS), such as nitric oxide—has been suggested to also contribute to age-related diseases, namely, hepatic steatosis and apoptosis [], as well as functional and structural changes in the cardiovascular system [, ], sleep homeostasis [], psychological disorders [], and dementia [].

Most supporters of the genomic instability theory of aging refer to telomere shortening [] and mutation in DNA mitochondrial. Telomeres are the repeated DNA sequences at the ends of linear chromosomes, which are unable to be fully replicated by DNA polymerases.

Mutations in mtDNA cause a wide range of human mitochondrial diseases and have been implicated in age-related diseases and aging.

Biomarker Features

Finding the biomarker of aging is one of the most important goals of medicine. The National Institutes of Health Biomarkers Definitions Working Group defined a biomarker as “a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention” [].

The American Federation for Aging Research (AFAR) recommends the following criteria for biomarkers of aging [•]:

  1. 1)

    It must predict a person’s physiological, cognitive, and physical function in an age-related way, independently of chronological age.

  2. 2)

    It must be testable and not harmful to test subjects (for example a blood test or an imaging technique); it must also be technically simple to perform, and it must be accurate and reproducibly without the need for specialized equipment or techniques.

  3. 3)

    It should work in laboratory animals as well as humans since preliminary testing is always done in nonhuman subjects.

Ferrucci et al. reviewed the biomarkers proposed as elements of a theory based on the balance between “resilience mechanisms” and “accumulated damages,” where biomarkers act in reducing resilience mechanisms or increasing damages [•] (Tables  and ).

Table 1 Biological changes underlying aging

Full size table

Table 2 Measurable biomarkers classified by respective hallmarks

Full size table

The pathways eligible to become biomarkers are the following:

Genomic Instability

Endogenous and exogenous agents continuously challenge the integrity of DNA; when DNA repair mechanisms cannot manage the repeated damage, the result is an accumulation of DNA somatic mutations. This phenomenon causes dysregulation of gene expression and the production of altered proteins that lead to cellular damage. Somatic mutation accumulation has been observed in skeletal muscle cells, neurons, and lymphocytes B related to aging [,,,]; nevertheless, quantification of DNA repair capacity in humans has yet to be finalized [,,].

Telomere Attrition

Telomeres are the DNA sequences that are placed at the end of the DNA chain and protect the chromosome ends from damage. During each replication, telomeres are reproduced, but not completely, so with aging they become shorter and contribute to cellular senescence [,,]. To date, different techniques are available to detect telomere length in circulating cells; however, no techniques have been validated for evaluating aging, because of the heterogeneity between different cells, between individuals and high measurement errors that make these techniques not yet valid in clinical practice [,,,].

Epigenetic Alterations

Epigenetics refers to those mechanisms, external to DNA, that modulate gene expression in cells; the regulation of gene expression determines the phenotypic characteristics of the different cells and tissues. The main mechanisms are DNA methylation, histone modification, and noncoding RNA. While DNA methylation is easily measured in circulating cells and seems to be correlated to aging [, ], measuring histone modification or noncoding RNA is difficult and expensive. Recent evidence correlates DNA methylation with aging and age-related chronic diseases in humans [, ]. Individuals with higher levels of DNA methylation have a higher risk of developing several age-related diseases and premature mortality for all causes and cardiovascular diseases [], as well as physical and cognitive functions [, ].

Loss of Proteostasis

The repair of damaged structures or their elimination is fundamental to maintain cell integrity and function []. Studies suggest that proteostasis becomes defective with aging and contributes to immunosenescence [] and that autophagy appears to be more functional in long-lived people []. Measuring the loss of proteostasis mechanism could be a good biomarker, but, to date, there are no valid techniques for this purpose.

Mitochondrial Dysfunction

The main role of mitochondria is to guarantee energy for the cell through the production of ATP. They are also involved in signaling by the production of ROS and in apoptosis-programmed cell death. Mitochondrial dysfunction is a good biomarker of aging and is associated with disability in older persons, through the reduction of muscle strength [•].

Many techniques are measuring oxidative phosphorylation and ROS generation that have been associated with chronic disease [, ]; nevertheless, the relation with aging is not completely validated.

Cellular Senescence

Genomic instability, telomere shortening, and other endogenous and exogenous mechanisms can induce the cell to activate specific pathways that lead to apoptosis []. This process is called cellular senescence and is characterized by structural and functional changes in the cell []. Senescent cells produce pro-inflammatory cytokines and chemokines, growth factors, and matrix proteases called “senescence-associated secretory phenotype” (SASP) [, ] which may induce some age-related diseases [,,]. The detection of SASP has been proposed as a biomarker of aging [].

Deregulated Nutrient-Sensing

Genetic mutations in growth hormone and the insulin-like growth factor have been linked to longevity []. Moreover, dietary restriction showed to increase life span in primates [, ]. For these reasons, this pathway has been proposed as biomarkers of aging.

Steam Cell Exhaustion

The decline in the regenerative potential is one of the elements at the base of aging []. Despite pharmacological interventions being explored to counteract this phenomenon [], evidences are still poor.

Altered Intercellular Communication

With aging, we also observe changes in intercellular communication: as inflammatory reaction increases, the other communication ways become dysfunctional (endocrine, neuronal, immune system) [].

As we discussed earlier, inflammation can be inappropriately increased in aging, and this has been related to age-related disease [, ].

Indeed, the pathways, described as potential biomarkers of aging, are strongly related to inflammation; for this reason, measuring circulating levels of cytokines is considered a new field of research [, •, ].

Aging and Life Expectancy

Aging and life expectancy are closely related. In a broad sense, determining an individual’s life expectancy is also a way of schematizing his or her aging process. Life expectancy is a statistical measure of the average time an organism is expected to live, based on the year of its birth (LEB), its current age and demographic factors including gender []. In the last decades, life expectancy has increased in high income country; the rise in human life expectancy has involved declines in intrinsic and extrinsic mortality processes associated, respectively, with senescence and environmental challenges [].

In association to this increased longevity, there are diseases called age-related that increase quadratically with age and cause a progressive loss of physical, mental, and cognitive integrities, leading to impaired function and increased vulnerability to morbidity, mortality [] and disability, in addition to increasing care needs and age-related burden measured through the sum of disability-adjusted life years (DALYs) of these diseases among these adults (Fig. ). Ninety-two of the 293 of the Global Burden of Disease causes were identified as age-related diseases. In particular, cardiovascular disease, neoplasm, and chronic respiratory disorders are those with higher age-related disease burden [].

Fig. 2

What is meant by biological aging?

Age-related diseases, adapted from Chang et al. []

Full size image

Determinants of Frailty Syndrome as Aging Biomarker

Frailty can be defined as a state of increased vulnerability to stressors or a loss of capacity to resolve homeostasis perturbation. Frailty condition is closely related to aging [••], and the frailty indexes can consequently be considered biomarkers of aging themselves. In frail individuals, it is possible to find both changing in body composition and balance between energy availability and energy demand. Moreover, in the definition of frailty, it is well described how signaling networks maintain homeostasis and association with neurodegeneration. These four aspects all refer to the hallmarks of aging. Frailty is associated with adverse clinical outcomes, including falls, institutionalization, and death [••].

Two principal models emerged in the last decades that are able to conceptualize and consequently measure frailty in everyday clinical practice and research: the “frailty phenotype” model and the cumulative deficits model.

The frailty phenotype was first described by Fried and colleagues in 2001, analyzing data from the Cardiovascular Health Study (CHS), involving 5210 men and women aged 65 years and older. In this study, it was investigated which characteristics of the population were predictive of falls, disability, hospitalization, and death. Their operational definition of frailty included a cluster of at least three of the following variables: unintentional weight loss, self-reported exhaustion, low energy expenditure, slow gait speed, and weak grip strength. This model does not take into consideration cognitive impairment as a cause of increased vulnerability, as this could contribute to functional decline and adverse events in older people [, ].

The cumulative deficits model was developed by Rockwood and colleagues as part of the prospective Canadian Study of Health and Aging (CSHA), involving a cohort of 10,263 older adults []. The authors identified 92 parameters, including diseases, disabilities, signs, symptoms, and laboratory values, which were defined as “deficits.” The sum of the deficits in a single individual allowed for the calculation of a frailty index (i.e., the number of deficits divided by 92). Frailty in this model is not considered as a cluster of symptoms but is conceptualized as a gradable syndrome, with a higher number of deficits implying an increased vulnerability state. The two models of frailty show significant overlap, although they capture slightly different sides of the same problem. It is important to notice that physical frailty is frequently associated with multimorbidity [, •, ].

It has been observed that the frailty phenotype construct is intrinsically related to mobility issues. Indeed, in older adults, physical performance measures are a robust and consistent predictor for disability, hospitalization, institutionalization, and death, both in the research and in the clinical setting. Lower physical performance is frequently associated with loss of skeletal muscle mass and quality, causing reduced strength and functional impairment [••]. This process has been called sarcopenia. Even though sarcopenia has been long associated with aging, it has to be acknowledged that it can develop much earlier in life []. Different definitions exist for this condition for the operational definition of sarcopenia both in the clinic and for research purposes that prioritize the assessment of muscle strength over muscle mass to identity sarcopenic patients. Strength is more closely related to survival and functional decline, compared with muscle mass [••]. According to EWGSOP criteria, sarcopenia is defined by the presence of low muscle strength (criterion 1) and either or low muscle quantity or quality (criterion 2) or low physical performance (criterion 3) [••].

The physical performance parameters used in the identification of frailty syndrome, both integrated (e.g. SPPB) and alone (walking speed, handgrip strength), can be used as aging performance biomarkers.

Determination of Medical and Social Needs

Why consider medical and social needs aging biomarkers?

In 1952, Robert J. Havighurst said: “In considering the needs of older people it is well, first, to remember that older people have the needs that are common to all people, and, second, that they have special needs due to the fact that they are old people”. This sentence describes everything there is to know about the need for the elderly and answers the question before.

In every society and age, there is what is meant by normality. An elderly person in this scenario needs what is needed to maintain this level of normalcy. Activity of daily living and instrumental activity of daily living (ADL and IADL) alone, remodelled according to the context and gender, can identify the minimum necessary. Conducting needs assessment, various areas must be considered including physical health, mental health, emotional, care, social, cultural, economic, nutritional, service, security, legal, and educational.

Many tools are used to evaluate people’s needs. The majority of these tools are focused on physical performance able to maintain autonomy; few studies focus on social needs and the costs of care. In the West World, 10% of patients account for 70% of total health care expenditures. This 10% is represented by older people, individuals with multiple chronic conditions, many medications, frequent hospitalizations, and limitations on their ability to perform basic daily functions due to physical, mental, or psychosocial challenge [].

Since the health care and social needs of older adults differ from that of other adults, it is necessary to identify the needs of the elderly to make proper plans that will promote their health.

Currently, most of the conducted studies had mainly focused on the elderly physical health needs and had neglected to take into account other needs such as social and health care needs. Furthermore, in addition to quantitative studies, discovering the older adults’ “perceptions” of their own health needs is also necessary.

Conclusion

There is a large interest of researchers in biomarkers of aging, and despite some of them seem to be very promising, biological biomarkers are still far from a clinical application; to date, there is no technique that meets the mentioned criteria of the ideal biomarker [•]. Moreover, we know that the biological pathways are the final agents of aging, but on one side they can be influenced by social, economic and environmental factors, and on the other side, they express in various disease and disabilities of the person (physical and cognitive impairments, age-related disease, systems functions, sensory functions, etc.) (Fig. ).

Fig. 3

What is meant by biological aging?

Mechanisms connecting different clusters of biomarkers

Full size image

To date, more than a single biomarker, to assess aging, we should consider a cluster of biomarkers that comprise the various elements that we analyzed: social and educational aspects, economic factors, country of origin, presence of age-related disease, presence of dependence in daily activities, physical capability, cognitive function, lung and cardiovascular function, and presence of sensory dysfunctions. In Table , we propose several clinical and laboratory biomarkers that can be used in clinical practice and research.

Table 3 Panel of possible biomarkers of aging

Full size table

The geriatric assessment (GA) can currently be considered a system capable of monitoring multiple biomarkers, clinical and laboratory, of aging, and at the same time able to relate them to each other. Through the GA, it is possible to make a prediction of the risk of toxicity of a treatment, of life expectancy, of social needs, and of compliance with the treatments. GA is composed, indeed, by several evaluations, made through standardized tools, which examine various aspects of the person (a multidimensional assessment).

Although it seems difficult to imagine a geriatric assessment as a biomarker, currently for its characteristics and for the high predictivity it has, it can be considered the gold standard in the management of the older individual and instrument toward which other biomarkers should be evaluated.

The purpose of this paper was to evaluate the multiple aspects that distinguish the aging process. Aging must no longer be described as a simple demographic event but as a complex mosaic in which several tesserae relate to each other, some in a very evident way others often in a more subdued but all fundamental way. Each aging theory has attempted to justify this process effectively; however, there is no single biomarker to date that has been found able to identify the stage of this process. At the same time, clinical clusters have been added to purely biological markers, and social ones should certainly be considered. It, therefore, becomes important not to consider biomarkers only as life span, but to try to overcome this link and focus on the set of factors that, influencing each other, are able to guide aging in good health and good quality of life towards a lived aging as a slow decline. At the time we are writing this paper, COVID 19 infection is reaping victims especially in Italy. The highest mortality is observed among the older adults, but surprisingly, it seems to maintain similar values between the youngest and oldest old (over 90 years). Currently, no plausible justification is provided for these data. In frailty, the number of comorbidities, the reduced functional reserve was the most used reasons. Indirectly, this infection is highlighting the need to use parameters that can more easily identify the aging process regardless of chronological age.

The studies analyzed in the literature show that if on the one hand there are physiological biomarkers able of highlighting some features of aging, other functional markers (performance, social and economic status, some pathologies and the presence of addiction) are able of speed it up or slow it down. For this reason, if we want to translate the use of biomarkers into clinical practice, we can think of not only something measurable through blood analysis but also a functional assessment of the patient we have in front, with his/her context and social network.

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. He W, Goodkind D, Kowal P. An aging world: 2015, International Population Reports. U.S. Government Printing Office, Washington DC. http://www.census.gov/library/publications/2016/demo/P95-16-1.html.

  2. Chang AY, Skirbekk VF, Tyrovolas S, Kassebaum NJ, Dieleman JL. Measuring population ageing: an analysis of the Global Burden of Disease Study 2017. The Lancet Public Health 2019;4(3):e159-e67.

  3. Colloca G, Tagliaferri L, Di Capua B, Gambacorta MA, Lanzotti V, Bellieni A, et al. Management of the elderly cancer patients complexity: the radiation oncology potential. Aging Dis. 2020;11(3):649e657.

  4. Cevenini E, Invidia L, Lescai F, Salvioli S, Tieri P, Castellani G, et al. Human models of aging and longevity. Expert Opin Biol Ther. 2008;8(9):1393–405.

    CAS  PubMed  Google Scholar 

  5. Colloca G, Santoro M, Gambassi G, Bernabei R. Aging and the management of related physiological changes. Geriatr Med Intell. 2009;18(2):67–78.

  6. Pinto Da Costa J, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T. A synopsis on aging-theories, mechanisms and future prospects. Ageing Res Rev. 2016;29:90–112.

  7. TBL K. Understanding the odd science of aging. Cell. Cell Press. 2005;120:437–47.

    Google Scholar 

  8. • Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, et al. Inflammaging and anti-inflammaging : a systemic perspective on aging and longevity emerged from studies in humans. Mech Ageing Dev. 2007;128:92–105. The aging phenotype, including immunosenescence, is explained by an imbalance between inflammatory and anti-inflammatory networks, which results in the low-grade chronic pro-inflammatory status that was proposed to call inflammaging by Franceschi. Inflammaging can be flanked by anti-inflammaging as major determinants of aging and longevity process.

    CAS  PubMed  Google Scholar 

  9. •• Colloca G, Di Capua B, Bellieni A, Cesari M, Valentini V, Marzetti E, et al. Muscoloskeletal aging, sarcopenia and cancer. J Geriatr Oncol. 2019;10(3):504–509. In this paper, it is described the correlation between sarcopenia and adverse outcomes such as treatment response or toxicity in cancer patients. The differences between the losses of muscle mass were linked to normal aging, an independent pathological condition such as sarcopenia and the cachexia.

  10. Lencel P, Magne D. Inflammaging: the driving force in osteoporosis? Med Hypotheses. 2011;76(3):317–21.

    CAS  PubMed  Google Scholar 

  11. Varadhan R, Yao W, Matteini A, Beamer BA, Xue QL, Yang H, et al. Simple biologically informed infammatory index of two serum cytokines predicts 10 year all-cause mortality in older adults. J Gerontol Ser A Biol Sci Med Sci. 2014;69 A(2):165–73.

    Google Scholar 

  12. Cevenini E, Monti D, Franceschi C. Inflamm-ageing. Curr Opin Clin Nutr Metab Care. 2013;16:14–20.

    CAS  PubMed  Google Scholar 

  13. Dollemore D. Aging under the microscope: a biological quest. Bethesda: National Institutes of Health National Institute on Aging Office of Communications and Public Liaison; 2002.

    Google Scholar 

  14. Goldberger AL, Amaral LAN, Hausdorff JM, Ivanov PC, Peng CK, Stanley HE. Fractal dynamics in physiology: alterations with disease and aging. Proc Natl Acad Sci U S A. 2002;99(SUPPL. 1):2466–72.

    PubMed  PubMed Central  Google Scholar 

  15. Kaplan RF, Verfaellie M, Meadows ME, Caplan LR, Pessin MS, Dewitt LD. Changing attentional demands in left hemispatial neglect. Arch Neurol. 1991;48(12):1263–6.

    CAS  PubMed  Google Scholar 

  16. Terrier P, Dériaz O. Kinematic variability, fractal dynamics and local dynamic stability of treadmill walking. J Neuroeng Rehabil. 2011;8:12.

  17. Schumann CM, Bloss CS, Barnes CC, Wideman GM, Carper RA, Akshoomoff N, et al. Longitudinal magnetic resonance imaging study of cortical development through early childhood in autism. J Neurosci. 2010;30(12):4419–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Azemin MZC, Kumar DK, Wong TY, Wang JJ, Mitchell P, Kawasaki R, et al. Age-related rarefaction in the fractal dimension of retinal vessel. Neurobiol Aging. 2012;33(1):194.e1–4.

    Google Scholar 

  19. Manor B, Costa MD, Kun H, Newton E, Starobinets O, Hyun GK, et al. Physiological complexity and system adaptability: evidence from postural control dynamics of older adults. J Appl Physiol. 2010;109(6):1786–91.

    PubMed  PubMed Central  Google Scholar 

  20. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013 [cited 2016 Oct 1];153(6):1194–217.

    PubMed  PubMed Central  Google Scholar 

  21. •• Franceschi C, Bonafè M, Valesin S. Inflamm-aging, An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244–54. In this paper, Franceschi argues that the persistence of inflammatory stimuli over time represents the biologic background favoring the susceptibility to age-related diseases/disabilities. A global reduction in the capacity to cope with a variety of stressors and a concomitant progressive increase in proinflammatory status are major characteristics of the aging process. This phenomenon was referred to as “inflammaging”.

    CAS  PubMed  Google Scholar 

  22. Goldsmith J, Crainiceanu CM, Caffo B, Reich D. Longitudinal penalized functional regression for cognitive outcomes on neuronal tract measurements. J R Stat Soc: Ser C: Appl Stat. 2012;61(3):453–69.

    Google Scholar 

  23. Johnson BL. Introduction to the special feature: adaptive management-scientifically sound, socially challenged. Conserv Ecol. 1999;3(1):10.

    Google Scholar 

  24. Aledo J, Blanco J. Aging is neither a failure nor an achievement of natural selection. Curr Aging Sci. 2015;8(1):4–10.

    PubMed  Google Scholar 

  25. Orr WC, Sohal RS. Extension of life-span by overexpression of superoxide dismutase and catalase in Drosophila melanogaster. Science (80- ). 1994;263(5150):1128–30.

    CAS  Google Scholar 

  26. Cui Z, Yang X, Shen Q, Wang K, Zhu T. Optimisation of biotransformation conditions for production of 2-phenylethanol by a Saccharomyces cerevisiae CWY132 mutant. Nat Prod Res. 2011;25(7):754–9.

    CAS  PubMed  Google Scholar 

  27. Artal-Sanz M, Tavernarakis N. Prohibitin couples diapause signalling to mitochondrial metabolism during ageing in C. elegans. Nature. 2009;461(7265):793–7.

    CAS  PubMed  Google Scholar 

  28. Afanas’ev I. New nucleophilic mechanisms of ROS-dependent epigenetic modifications: comparison of aging and cancer. Aging Dis. 2014;5(1):52–62.

    PubMed  Google Scholar 

  29. Thanan R, Oikawa S, Hiraku Y, Ohnishi S, Ma N, Pinlaor S, et al. Oxidative stress and its significant roles in neurodegenerative diseases and cancer. Int J Mol Sci. MDPI AG. 2014;16:193–217.

    PubMed  PubMed Central  Google Scholar 

  30. Nyström T. Role of oxidative carbonylation in protein quality control and senescence. EMBO J. 2005;24:1311–7.

    PubMed  PubMed Central  Google Scholar 

  31. Abdelmegeed MA, Choi Y, Ha SK, Song BJ. Cytochrome P450-2E1 is involved in aging-related kidney damage in mice through increased nitroxidative stress. Food Chem Toxicol. 2017;109:48–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Novella S, Dantas AP, Segarra G, Novensa L, Heras M, Hermenegildo C, et al. Aging enhances contraction to thromboxane A2 in aorta from female senescence-accelerated mice. Age (Omaha). 2013;35(1):117–28.

    CAS  Google Scholar 

  33. Surikow SY, Raman B, Licari J, Singh K, Nguyen TH, Horowitz JD. Evidence of nitrosative stress within hearts of patients dying of Tako-tsubo cardiomyopathy. Int J Cardiol. 2015;189(1):112–4.

    CAS  PubMed  Google Scholar 

  34. Rytkönen KM, Wigren HK, Kostin A, Porkka-Heiskanen T, Kalinchuk AV. Nitric oxide mediated recovery sleep is attenuated with aging. Neurobiol Aging. 2010;31(11):2011–9.

    PubMed  Google Scholar 

  35. Maurya PK, Noto C, Rizzo LB, Rios AC, Nunes SOV, Barbosa DS, et al. The role of oxidative and nitrosative stress in accelerated aging and major depressive disorder. Prog Neuro-Psychopharmacol Biol Psychiatry. Elsevier Inc. 2016;65:134–44.

    CAS  Google Scholar 

  36. Mangialasche F, Polidori MC, Monastero R, Ercolani S, Camarda C, Cecchetti R, et al. Biomarkers of oxidative and nitrosative damage in Alzheimer’s disease and mild cognitive impairment. Ageing Res Rev. 2009;8:285–305.

    CAS  PubMed  Google Scholar 

  37. Kruk PA, Rampino NJ, Bohr VA. DNA damage and repair in telomeres: relation to aging. Proc Natl Acad Sci U S A. 1995;92(1):258–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Atkinson AJ, Colburn WA, DeGruttola VG, DeMets DL, Downing GJ, Hoth DF, et al. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69:89–95.

    Google Scholar 

  39. • Biomarkers of aging an introduction to aging science brought to you by the American Federation for Aging Research, info aging guides. 2016.

  40. • Ferrucci L, Gonzalez-Freire M, Fabbri E, et al. Measuring biological aging in humans: A quest. Aging Cell. 2020;19:e13080.

  41. Zhang L, Dong X, Lee M, Maslov AY, Wang T, Vijg J. Single-cell whole-genome sequencing reveals the functional landscape of somatic mutations in B lymphocytes across the human lifespan. Proc Natl Acad Sci U S A. 2019;116(18):9014–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Franco I, Johansson A, Olsson K, Vrtačnik P, Lundin P, Helgadottir HT, et al. Somatic mutagenesis in satellite cells associates with human skeletal muscle aging. Nat Commun. 2018;9(1):800.

  43. Bae T, Tomasini L, Mariani J, Zhou B, Roychowdhury T, Franjic D, et al. Different mutational rates and mechanisms in human cells at pregastrulation and neurogenesis. Science (80- ). 2018;359(6375):550–5.

    CAS  Google Scholar 

  44. Lodato MA, Rodin RE, Bohrson CL, Coulter ME, Barton AR, Kwon M, et al. Aging and neurodegeneration are associated with increased mutations in single human neurons. Science (80- ). 2018;359(6375):555–9.

    CAS  Google Scholar 

  45. Berwick M, Vineis P. Measuring DNA repair capacity: small steps. J Natl Cancer Inst. 2005;97(2):84–5.

    PubMed  Google Scholar 

  46. Trzeciak AR, Barnes J, Ejiogu N, Foster K, Brant LJ, Zonderman AB, et al. Age, sex, and race influence single-strand break repair capacity in a human population. Free Radic Biol Med. 2008;45(12):1631–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Trzeciak AR, Barnes J, Evans MK. A modified alkaline comet assay for measuring DNA repair capacity in human populations. Radiat Res. 2008;169(1):110–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Greider CW. Telomerase discovery: the excitement of putting together pieces of the puzzle (Nobel lecture). Angew Chem Int Ed. 2010;49:7422–39.

    CAS  Google Scholar 

  49. Saretzki G. Telomeres, telomerase and ageing. In: Subcellular biochemistry. New York: Springer; 2018. p. 221–308.

    Google Scholar 

  50. Vera E, Bernardes de Jesus B, Foronda M, Flores JM, Blasco MA. The rate of increase of short telomeres predicts longevity in mammals. Cell Rep. 2012;2(4):732–7.

    CAS  PubMed  Google Scholar 

  51. Berglund K, Reynolds CA, Ploner A, Gerritsen L, Hovatta I, Pedersen NL, et al. Longitudinal decline of leukocyte telomere length in old age and the association with sex and genetic risk. Aging (Albany NY). 2016;8(7):1398–415.

    CAS  Google Scholar 

  52. Bischoff C, Petersen HC, Graakjaer J, Andersen-Ranberg K, Vaupel JW, Bohr VA, et al. No association between telomere length and survival among the elderly and oldest old. Epidemiology. 2006;17(2):190–4.

    PubMed  Google Scholar 

  53. Lin J, Cheon J, Brown R, Coccia M, Puterman E, Aschbacher K, et al. Systematic and cell type-specific telomere length changes in subsets of lymphocytes. J Immunol Res. 2016;2016:1–9.

    Google Scholar 

  54. Müezzinler A, Zaineddin AK, Brenner H. A systematic review of leukocyte telomere length and age in adults. Ageing Res Rev. 2013;12(2):509–19.

    PubMed  Google Scholar 

  55. Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda SV, et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013;49(2):359–67.

    CAS  PubMed  Google Scholar 

  56. Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(10):3156.

  57. Gensous N, Bacalini MG, Pirazzini C, Marasco E, Giuliani C, Ravaioli F, et al. The epigenetic landscape of age-related diseases: the geroscience perspective. Biogerontology. 2017;18(4):549–59.

    PubMed  PubMed Central  Google Scholar 

  58. Levine ME, Lu AT, Quach A, Chen BH, Assimes TL, Bandinelli S, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 2018;10(4):573–91.

    Google Scholar 

  59. Chen BH, Marioni RE, Colicino E, Peters MJ, Ward-Caviness CK, Tsai PC, et al. DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging (Albany NY). 2016;8(9):1844–65.

    CAS  Google Scholar 

  60. Degerman S, Josefsson M, Nordin Adolfsson A, Wennstedt S, Landfors M, Haider Z, et al. Maintained memory in aging is associated with young epigenetic age. Neurobiol Aging. 2017;55:167–71.

    PubMed  Google Scholar 

  61. Gale CR, Marioni RE, Čukić I, Chastin SF, Dall PM, Dontje ML, et al. The epigenetic clock and objectively measured sedentary and walking behavior in older adults: the Lothian Birth Cohort 1936. Clin Epigenetics. 2018;10(1):4.

    PubMed  PubMed Central  Google Scholar 

  62. Cuervo AM, Bergamini E, Brunk UT, Dröge W, Ffrench M, Terman A. Autophagy and aging: the importance of maintaining “clean” cells. Autophagy. 2005;1(3):131–40.

    PubMed  Google Scholar 

  63. Cuervo AM, Macian F. Autophagy and the immune function in aging. Curr Opin Immunol. 2014;29(1):97–104.

    CAS  PubMed  Google Scholar 

  64. Raz Y, Guerrero-Ros I, Maier A, Slagboom PE, Atzmon G, Barzilai N, et al. Activation-induced autophagy is preserved in CD4+ T-cells in familial longevity. J Gerontol A Biol Sci Med Sci. 2017;72(9):1201–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. • Zane AC, Reiter DA, Shardell M, Cameron D, Simonsick EM, Fishbein KW, et al. Muscle strength mediates the relationship between mitochondrial energetics and walking performance. Aging Cell. 2017;16(3):461–8. Impaired oxidative capacity affects muscle performance and, through this mechanism, has a negative effect on walking speed in the Baltimore Longitudinal Study of Aging. This is the first demonstration in human adults that mitochondrial function affects muscle strength and that inefficiency in musclebioenergetics partially accounts for differences in mobility through this mechanism.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Dikalov SI, Harrison DG. Methods for detection of mitochondrial and cellular reactive oxygen species. Antioxid Redox Signal. 2014;20(2):372–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Gonzalez-Freire M, Scalzo P, D’Agostino J, Moore ZA, Diaz-Ruiz A, Fabbri E, et al. Skeletal muscle ex vivo mitochondrial respiration parallels decline in vivo oxidative capacity, cardiorespiratory fitness, and muscle strength: the Baltimore Longitudinal Study of Aging. Aging Cell. 2018;17(2):e12725.

    PubMed Central  Google Scholar 

  68. Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, et al. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov. 2017;16(10):718–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15(7):482–96.

    PubMed  Google Scholar 

  70. Andriani GA, Almeida VP, Faggioli F, Mauro M, Li Tsai W, Santambrogio L, et al. Whole chromosome instability induces senescence and promotes SASP. Sci Rep. 2016;6:35218.

  71. Coppé J-P, Desprez P-Y, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol Mech Dis. 2010;5(1):99–118.

    Google Scholar 

  72. Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest. 2018;128(4):1208–16.

    PubMed  PubMed Central  Google Scholar 

  73. Palmer AK, Tchkonia T, LeBrasseur NK, Chini EN, Xu M, Kirkland JL. Cellular senescence in type 2 diabetes: a therapeutic opportunity. Diabetes. 2015;64(7):2289–98.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Waters DW, Blokland KEC, Pathinayake PS, Burgess JK, Mutsaers SE, Prele CM, et al. Fibroblast senescence in the pathology of idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2018;315(2):L162–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Tanaka T, Biancotto A, Moaddel R, Moore AZ, Gonzalez-Freire M, Aon MA, et al. Plasma proteomic signature of age in healthy humans. Aging Cell. 2018;17(5):e12799.

    PubMed  PubMed Central  Google Scholar 

  76. Barzilai N, Huffman DM, Muzumdar RH, Bartke A. The critical role of metabolic pathways in aging. Diabetes. 2012;61(6):1315–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Mattison JA, Roth GS, Mark Beasley T, Tilmont EM, Handy AM, Herbert RL, et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature. 2012;489(7415):318–21.

    CAS  PubMed  Google Scholar 

  78. Anderson RM, Le Couteur DG, de Cabo R. Caloric restriction research: new perspectives on the biology of aging. J Gerontol A Biol Sci Med Sci. 2018;73(1):1–3.

    Google Scholar 

  79. Ren R, Ocampo A, Liu GH, Izpisua Belmonte JC. Regulation of stem cell aging by metabolism and epigenetics. Cell Metab. 2017;26(3):460–74.

    CAS  PubMed  Google Scholar 

  80. Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell. 2012;148(1–2):46–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Russell SJ, Kahn CR. Endocrine regulation of ageing. Nat Rev Mol Cell Biol. 2007;8(9):681–91.

    CAS  PubMed  Google Scholar 

  82. Bektas A, Schurman SH, Sen R, Ferrucci L. Aging, inflammation and the environment. Exp Gerontol. 2018;105:10–8.

    CAS  PubMed  Google Scholar 

  83. Fabbri E, An Y, Zoli M, Simonsick EM, Guralnik JM, Bandinelli S, et al. Aging and the burden of multimorbidity: associations with inflammatory and anabolic hormonal biomarkers. J Gerontol A Biol Sci Med Sci. 2015;70(1):63–70.

    CAS  PubMed  Google Scholar 

  84. • Ferrucci L, Harris TB, Guralnik JM, Tracy RP, Corti M-C, Cohen HJ, et al. Serum IL-6 level and the development of disability in older persons. J Am Geriatr Soc. 1999;47(6):639–46. The serum concentration of interleukin 6 (IL-6), a cytokine that plays a central role in inflammation, increases with age. Higher circulating levels of IL-6 predict disability onset in older persons. This may be attributable to a direct effect of IL-6 on muscle atrophy and/or to the pathophysiologic role played by IL-6 in specific diseases.

    CAS  PubMed  Google Scholar 

  85. Ferrucci L, Penninx BWJH, Volpato S, Harris TB, Bandeen-Roche K, Balfour J, et al. Change in muscle strength explains accelerated decline of physical function in older women with high interleukin-6 serum levels. J Am Geriatr Soc. 2002;50(12):1947–54.

    PubMed  Google Scholar 

  86. Booth H, Tickle L. Mortality modelling and forecasting: a review of methods. 2008.

  87. Siegel JS. The demography and epidemiology of human health and aging. Dordrecht: Springer Netherlands; 2012. p. 1–985.

    Google Scholar 

  88. •• Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. Frailty in elderly people. Lancet. 2013;381(9868):752–62. Frailty is the most problematic expression of population aging. It is a state of vulnerability to poor resolution of homoeostasis after a stressor event and is a consequence of cumulative decline in many physiological systems during a lifetime. This cumulative decline depletes homoeostatic reserves until minor stressor events trigger disproportionate changes in health status.

    Google Scholar 

  89. Avila-Funes JA, Amieva H, Barberger-Gateau P, Le Goff M, Raoux N, Ritchie K, et al. Cognitive impairment improves the predictive validity of the phenotype of frailty for adverse health outcomes: the three-city study. J Am Geriatr Soc. 2009;57(3):453–61.

    PubMed  Google Scholar 

  90. La Carpia D, Liperoti R, Guglielmo M, Di Capua B, Devizzi LF, Matteucci P, et al. Cognitive decline in older long-term survivors from non-Hodgkin lymphoma: a multicenter cross-sectional study. J Geriatr Oncol. 2020;11(5):790–795.

  91. Rockwood K, Mitnitski A. Frailty in relation to the accumulation of deficits. J Gerontol A Biol Sci Med Sci. 2007;62(7):722–7.

    PubMed  Google Scholar 

  92. Hanlon P, Nicholl BI, Jani BD, Lee D, McQueenie R, Mair FS. Frailty and pre-frailty in middle-aged and older adults and its association with multimorbidity and mortality: a prospective analysis of 493 737 UK Biobank participants. Lancet Public Health. 2018;3(7):e323–32.

    PubMed  PubMed Central  Google Scholar 

  93. • Fried LP, Ferrucci L, Darer J, Williamson JD, Anderson G. Untangling the concepts of disability, frailty, and comorbidity: implications for improved targeting and care. J Gerontol A Biol Sci Med Sci. 2004;59(3):M255–63. This provides a basis for distinguishing between these three important clinical conditions in older adults (disability, frailty, comorbidity) and showing how use of separate, distinct definitions of each can improve our understanding of the problems affecting older patients and lead to development of improved strategies for diagnosis, care, research and medical education in this area.

    Google Scholar 

  94. Galli E, Cuccaro A, Maiolo E, Bellesi S, D’Alò F, Fusco D, et al. Comorbidity assessment to determine prognosis in older adult patients with classical Hodgkin lymphoma. Hematol Oncol. 2020;38(2):153–161.

  95. •• Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48(4):601. European consensus on sarcopenia definition and diagnosis.

    PubMed  PubMed Central  Google Scholar 

  96. Sayer AA, Syddall HE, Gilbody HJ, Dennison EM, Cooper C. Does sarcopenia originate in early life? Findings from the Hertfordshire cohort study. J Gerontol A Biol Sci Med Sci. 2004;59(9):M930–4.

    PubMed  Google Scholar 

  97. Bodenheimer T, Berry-Millett R. Follow the money--controlling expenditures by improving care for patients needing costly services. N Engl J Med. 2009;361(16):1521–3.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

  1. U.O.C. di Radioterapia Oncologica, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy

    Giuseppe Colloca, Beatrice Di Capua, Luca Tagliaferri & Vincenzo Valentini

  2. Dipartimento di Scienze dell’invecchiamento, neurologiche, ortopediche e della testa-collo, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy

    Andrea Bellieni, Domenico Fusco & Francesca Ciciarello

  3. Moffitt Cancer Center, Tampa, FL, USA

    Lodovico Balducci

Authors

  1. Giuseppe Colloca

    View author publications

    You can also search for this author in PubMed Google Scholar

  2. Beatrice Di Capua

    View author publications

    You can also search for this author in PubMed Google Scholar

  3. Andrea Bellieni

    View author publications

    You can also search for this author in PubMed Google Scholar

  4. Domenico Fusco

    View author publications

    You can also search for this author in PubMed Google Scholar

  5. Francesca Ciciarello

    View author publications

    You can also search for this author in PubMed Google Scholar

  6. Luca Tagliaferri

    View author publications

    You can also search for this author in PubMed Google Scholar

  7. Vincenzo Valentini

    View author publications

    You can also search for this author in PubMed Google Scholar

  8. Lodovico Balducci

    View author publications

    You can also search for this author in PubMed Google Scholar

Corresponding author

Correspondence to Beatrice Di Capua.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Geriatric Oncology

Rights and permissions

Reprints and Permissions

About this article

Verify currency and authenticity via CrossMark

Cite this article

Colloca, G., Di Capua, B., Bellieni, A. et al. Biological and Functional Biomarkers of Aging: Definition, Characteristics, and How They Can Impact Everyday Cancer Treatment. Curr Oncol Rep 22, 115 (2020). https://doi.org/10.1007/s11912-020-00977-w

What biologically causes aging?

Researchers continue to identify and explain key factors affecting the rate of aging, including the body's response to a variety of stresses; the function of the immune system; the role of cellular senescence; the body's response to macromolecular damage (damage to large molecules such as proteins or lipids), such as ...

What is an example of biological age?

Research on Methylation and Biological Age For example, healthy breast tissue can be as much as three years older than the rest of your body. If it's next to cancerous tissue, it's an average of 12 years older. Sex exists on a continuum.