Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 1.
Published in final edited form as: Curr Opin Psychiatry. 2019 Sep;32(5):381–387. doi: 10.1097/YCO.0000000000000525

Accelerated Aging in Serious Mental Disorders

F Saverio Bersani (1),(2), Synthia H Mellon (3), Victor I Reus (2), Owen M Wolkowitz (2),*
PMCID: PMC6707774  NIHMSID: NIHMS1534084  PMID: 31145144

Abstract

Purpose of Review:

Clinical, epidemiological and biological evidence raise the possibility that serious mental disorders (SMD’s) are associated with accelerated biological aging. To the extent this is true, SMD’s should not simply be considered in terms of mental illness or brain dysfunction, but also as “whole body” and multi-system illnesses, or else as conditions with significant somatic concomitants.

Recent Findings:

The concept of accelerated biological aging in SMD’s is supported by reports of accelerated changes in certain biomarkers normally associated with the aging process.

Summary:

We define and discuss several proposed biological aging markers that have been examined in SMD’s, we review the most recent findings, and we conclude with opinions regarding the merits and meanings of these markers, their usefulness in understanding and treating SMD’s, and remaining questions and future directions in this area of research.

Keywords: Serious mental disorders, Aging, Biological aging, Telomeres, Epigenetics

1. INTRODUCTION

Serious mental disorders (SMD’s) are associated with an increased risk of medical illnesses and premature mortality from natural causes, with lifespans up to 25 years shorter than the general population [1]. Although lifestyle and socioeconomic factors play a role, the psychiatric condition itself may be an independent risk factor, even after excluding death by suicide [1]. The particular medical illnesses that are more frequent in SMD’s are those that are more commonly seen with advanced age, such as cardiovascular disease and others. This has raised the possibility that SMD’s are associated with accelerated biological aging. Whereas chronological age is measured by the passage of time, biological age is defined physiologically and functionally and is more closely associated with disease processes and mortality.

2. PROPOSED MARKERS OF BIOLOGICAL AGING IN PSYCHIATRY

2.1. Telomere Length

Telomere length (TL) determinations, generally in peripheral leukocytes (LTL), are the most widely studied markers of biological aging in SMD’s. Telomeres, which cap DNA strands, protect chromosomes from damage and replicative senescence [2]. Telomeres shorten with repeated mitoses as well as with chronic exposure to oxidation, inflammation and possibly to the stress hormones, cortisol and catecholamines, unless acted upon by the telomere-lengthening enzyme, telomerase, or by alternative telomere-lengthening mechanisms [2]. When telomeres critically shorten, cells undergo replicative senescence or apoptosis or become genomically unstable. Telomere length inversely tracks chronological age, as LTL’s shorten at an average rate of approximately 25–30 base pairs per year, and the correlation between LTL and chronological age has been reported as −0.30 [3]. Shortened LTL is associated with, and longitudinally predicts, poor physical health and is significantly correlated with all-cause mortality [46]. Most studies have replicated findings of LTL shortening in chronic psychological stress and in SMD’s (especially major depressive disorder - MDD) [2,5,78], but the “toxic ingredients” of stress and SMD are unknown. Increases in inflammation and oxidative stress and stress hormones are prime candidates [2,813]. Because these biochemical factors can, themselves, be associated with physical disease and decreased life span, it is uncertain whether LTL shortening directly relates to health and age-associated outcomes, or rather, serves as a proxy or “canary in a coal mine,” informing on a toxic cellular milieu [2,14], and these possibilities are not mutually exclusive [15]. In any event, it is possible that LTL shortening has pathophysiologic significance in its own right [2,14].

Further complicating interpretation of telomere shortening in SMD’s is the question of which comes first. Studies on within-person longitudinal relationships between LTL and certain psychiatric symptoms (mainly symptoms of depression and anxiety), which could suggest a causal direction, have provided so far contrasting findings [1621]. While it is intuitive to assume that stress and SMD’s eventuate in shortened telomeres, it is also possible that (a) shortened telomeres are a risk factor for developing certain SMD’s [2225], and (b) shortened telomeres and SMD’s both arise from common antecedents [15,26] such as environmental factors, or common genetic underpinnings. For example, first episode, never-medicated depressed adolescents already show significant LTL shortening [23], as do never-depressed girls at high risk for developing depression [27]. Indeed, familial risk for MDD [27] or bipolar disorder (BD) [28] are reportedly associated with reduced LTL, although a recent cohort study showed that genetic risk for MDD, BD and schizophrenia was not associated with shorter LTL [29]. Telomere length is partly heritable, with estimates of 64% at baseline and 28% for rates of attrition [30], but heredity interacts with the environment in predicting TL [15]. Several GWAS studies have identified single nucleotide polymorphisms (SNPs) showing associations with LTL [3137]; one study found that a specific genetic variation of TERT (rs2736100), the catalytic subunit of telomerase, was associated with certain types of clinical depression [38], but this was not replicated by Michalek et al. [39], who instead found that a variant in the TERC gene (rs10936599), coding for the RNA component of telomerase, predicted increased risk for childhood-onset MDD, albeit accounting for only 3% of the variance. Moreover, several recent reports have not shown significant associations between genetic predisposition to shorter LTL and the risk of clinically significant depression [4041], including the largest study yet completed (N=67,306) [42]. Thus far, failures in replication, as well as issues of population stratification and limitations of the candidate gene approach in general, make any specific genetic association to psychiatric syndromes highly speculative.

2.2. Epigenetic Aging

More recently described markers, based on methylation of the genome, may provide even stronger estimates of biological age [43]. Age-associated site-specific methylation changes occur with surprising regularity across individuals and in some cases across tissues. Assessing such changes at specific 5’-C-phosphate-G-3’ (CpG) sites can indicate “DNA methylation age” or “epigenetic age” (EpiAge). Correlations between EpiAge and chronological age are remarkably high, with correlations of up to 0.96 [44]. Advanced epigenetic age is associated with many serious medical illnesses and predicts mortality better than chronological age alone [45]. Several different measures of epigenetic aging (termed “clocks”) have been developed. While each is strongly associated with chronological age and certain illnesses and mortality, each has specific properties and meanings. The Horvath clock was the first developed [44] and is based on methylation patterns of a set of 353 CpG’s (out of 21,369 examined) that was “trained” on predicting chronological age and then validated in independent samples; it was found to not only accurately predict chronological age, but to be more strongly associated with biological aging parameters. Soon thereafter, the Hannum clock was developed, based on a largely non-overlapping set of 71 CpG’s [46]. The Horvath clock assesses “intrinsic epigenetic aging” (IEAA), which is irrespective of cell or tissue type (and specifically controls for differences in leukocyte subpopulations), whereas the Hannum clock assesses “extrinsic epigenetic aging” (EEAA), which incorporates information about leukocyte sub-populations that also change with aging. Han et al used a newer DNA methylation algorithm that examined virtually the entire 28 million CpG sites to assess epigenetic aging in MDD [47] and found a set of 80,000 CpG sites that revealed a modest but significant acceleration of EpiAge in MDD. Notably, pathway analysis of the top CpG sites associated with epigenetic aging in MDD implicated neurogenesis, neuron differentiation and regulation of neuron death [47]. The only study to examine blood-based epigenetic aging in BD found no overall difference in EpiAge, although accelerated EpiAge was reported in the older BD subjects in that study [48]. Studies in schizophrenia mainly did not show show accelerated EpiAge in blood or brain samples [4952]. Epigenetic aging has been more extensively studied in PTSD or lifetime stress. Lifetime stress in urban African-Americans was associated with accelerated EpiAge, possibly secondary to glucocorticoid activation [53]. Individuals exposed to combat trauma showed accelerated EpiAge, but, paradoxically, those who developed PTSD showed an attenuation of this acceleration [5455]. Perhaps related to this paradox, cases of PTSD showed EpiAging in direct proportion to ratings of resiliency [56], possibly secondary to compensatory processes such as telomerase activation [54]. In a separate study, overall lifetime PTSD severity was not associated with EpiAge changes, but the lifetime severity of the cluster of PTSD symptoms related to hyper-arousal was associated with an acceleration of EpiAge [57]. A recent meta-analysis found that lifetime PTSD symptom severity was associated with advanced Hannum (but not Horvath) EpiAge [58], but a later longitudinal study found that baseline PTSD symptoms of avoidance and numbing predicted subsequent accelerated EpiAge by the Horvath, but not the Hannum, clock [59].

While TL and these epigenetic clocks all significantly correlate with chronological age and predict disease and mortality [6], they are independent from each other [60], and their mediators likely differ, with TL mostly affected by repeat mitoses, inflammation and oxidative stress [2], and certain epigenetic clocks possibly affected by glucocorticoids, age-related variations of methylcytosine or increasing age-related entropy of the methylome [53,6163] and, in the case of the Hannum clock, inflammation [6465]. Interestingly, TERT, which is generally associated with longer TL, may paradoxically confer higher IEAA [66]. Nonetheless, both TL and methylation are affected by the environment and lifestyle behaviors (e.g., sleep, diet, smoking and exercise) [26,62,67], which has obvious clinical implications.

Other recently introduced epigenetic clocks also correlate strongly with chronological age, but they more strongly predict disease and mortality; however, these promising new measures have yet to be examined in SMD’s. Methylation profiles of these latter clocks were developed specifically to predict lifespan and health span rather than chronological age alone. These clocks were trained on, in addition to age, clinical laboratory measures (“phenotypic age”) that generally change with aging and that predict illness and mortality (e.g., albumin, creatinine, glucose, C-reactive protein, white blood cell count and others), called “DNAm PhenoAge” [68], or on a selection of plasma proteins that have previously been associated with mortality or morbidity (e.g., plasminogen activator inhibitor-1 [PAI-1], cystatin C, leptin and others) as well as on methylation changes related to cigarette smoking history, called “DNAm GrimAge” [67]. This latter clock reportedly strongly predicts time-to-death. Of note, several of the factors implicated in DNAm GrimAge are also associated with shortened LTL [69].

At this early stage of investigation, caution must be exercised in interpreting findings of epigenetic aging in SMD’s, because of the small sample sizes and/or small effect sizes or hazard ratios in several of the studies [67], various technical challenges, uncertain interpretations of the different measures of EpiAge (and their modest inter-correlations [67]), and inadequate deep phenotyping of the subjects [65]. In particular, use of psychotropic or other medications, or the presence of comorbid medical or psychiatric conditions or tobacco/substance use, could instill major confounds into many of these studies.

2.3. Emerging Markers of Accelerated Aging

2.3a. Mitochondria

Mitochondrial dysfunction may reflect, and perhaps also play a role in, accelerated biological aging [7071] and is being studied in certain SMD’s. The relationships between telomere shortening and other indices of aging, such as mitochondrial dysfunction and its associated consequences of impaired oxidative metabolism, especially in SMD’s, are complex and remain incompletely understood [19,70,7281]. The literature may be inconsistent in part because mitochondrial status is variably defined by parameters of structure, copy number and function, which can change in relationship to each other over time [76,79] and can differ in different cellular subtypes [8283], and because one mitochondrial parameter in particular, mitochondrial DNA copy number, may bear an “inverted-U”-shaped relationship with cellular health [72,8485]. Evidence for a causal mechanistic connection between TL and age-associated mitochondrial parameters exists in both directions [8692]. The relationship between mitochondrial parameters and EpiAge is also of interest. D’Aquila et al [93] performed methylation analyses on CpG sites in candidate genes associated with mitochondria quality control and identified two genes (RAB32; RHOT2), confirmed by replication, that regulated mitochondrial aging. Interestingly, higher methylation levels in RHOT2 predicted greater disability in the subject population.

2.3b. Immunosenescence/ Inflammaging

Ageing-associated systemic, low-grade inflammation, termed “inflammaging,” is characterized by chronically increased levels of inflammatory cytokines and acute phase reactants and may underlie the progression of pathological senescence processes, including those in the brain [9496]. While chronic low level inflammation has repeatedly been demonstrated in MDD and various other SMD’s (at least in a subset of such patients) [97], its role in accelerating biological aging and its utility as a biomarker biological aging [98102] have yet to be adequately studied in SMD’s. A novel biological aging marker called “IMM-AGE” takes into consideration the relative abundance of 33 immune cell subsets that are consistently associated with age as well as with the function of these cells to express certain genes and to produce and react to cytokines [103]. This measure, which has yet to be assessed in SMD’s, reportedly correlates with overall survival more than 500-fold better than does than the Horvath EpiAge clock and may be more accurate for assessing overall all-cause mortality risk.

3. CONCLUSION

The landscape of SMD’s is changing, with a new focus on subcellular components and processes in addition to neurotransmitters. To the extent accelerated biological aging occurs in SMD’s, the scope of their pathophysiology broadens considerably, and they might no longer be framed as only “mental disorders” or even brain diseases, but rather as whole-body, multi-system illnesses (or at least as illnesses with substantial somatic comorbidity), of which the psychiatric presentation is just the most readily observable pathology [8,104]. This should lead to improved targeting of specific underlying pathologies (“personalized medicine”).

Challenges in appraising the significance of these biomarkers include:

  • Elucidating the relevance of peripheral blood biomarkers to the brain and other somatic cells;

  • Understanding whether the biomarkers have diagnostic specificity, or rather, are related to underlying trans-diagnostic physiological processes, including those suggested by the NIMH Research Domain Criteria (RDoC) [105]; inflammation, oxidative stress and glucocorticoids seem particularly relevant to several of the markers reviewed here [13,96,106109];

  • Differentiating “accelerated” from “premature” aging [110];

  • Clarifying whether the biomarkers are causally related to the aging process or merely epiphenomena.

Biological aging is likely a multi-faceted process, not easily quantifiable by a single biomarker [111112]. The analogy of the six blind men describing an elephant as a snake, a tree trunk, a broad leaf, etc., seems especially pertinent in defining biological aging. Already, we are seeing different calibrating tools that measure different aspects of aging, such as TL, mitochondrial functioning, immune activation, epigenetic aging and others. With the advent of powerful ‘omics tools, massive amounts of data can be collected and “trained” against different markers (e.g., chronological age, disability, disease, clinical lab test values, time to death, etc.), to provide different types of information.

Perhaps the greatest value of biomarkers of aging is their therapeutic utility. Among the most important questions is whether biological aging in SMD’s can be decelerated with appropriate interventions [2,113114]. Behavioral and lifestyle interventions can likely attenuate the pace of certain types of biological aging [115117]. Preliminary evidence also suggests that certain pharmacological therapies may retard biological aging [113], although few prospective, double-blind trials have yet been conducted. In our opinion, cross-sectional testing of aging biomarkers for clinical purposes is not “ready for prime time,” due to differences in assay techniques, lack of normative ranges and lack of knowledge about the effect of covariates. If anything, biomarker testing may be useful in longitudinal tracking within individuals to assess trajectories of these markers and possibly to indicate whether therapeutic interventions are called for and are effective [43,118119]. In all, we expect that understanding aging markers in SMD’s will accelerate our diagnostic and therapeutic approaches to these conditions and help clarify their pathophysiologies.

KEY BULLET POINTS:

  • Biological aging differs from chronological aging and is more closely related to serious age-related illnesses and mortality.

  • Serious mental illnesses may be characterized by accelerated biological aging.

  • Accelerated biological aging may contribute to increased illness and mortality in individuals with serious mental illnesses.

  • Telomere shortening, epigenetic changes, glucocorticoids and “inflammaging” may contribute to accelerated biological aging.

  • Lifestyle as well as pharmacological interventions may have the potential to decelerate the pace of biological aging.

ACKNOWLEDGEMENTS:

1. We would like to thank many colleagues and collaborators who have worked with us in investigating biological aging in SMD’s: Daniel Lindqvist, Jee In Kang, Elissa Epel, Aric Prather, Christina Hough, Laura Mahan, Rebecca Rosser, Heather Burke, Ryan Rampersaud, Brent Nier, Gwyneth Winnie Wu, Mina Cheema, Allie Morford, Jue Lin, Elizabeth Blackburn, Firdaus Dhabhar, Felipe Jain, Charles Marmar, Rachel Yehuda, Ruoting Yang, Rasha Hammamieh, Marti Jett, Maryam Khan, Åsa Westrin, Johan Fernström, Kristoffer Månnson, John Kelsoe, Yabin Wei, Josine Verhoeven, Brenda Penninx, Dora Réveész, Tony Yang, Olga Tymofiyeva, Eva Henje-Blom, Dilip Jeste, Sabrina Darrow, Kirsten Aschbacher, Aoife O’Donovan, Susanne Mueller, Dieter Meyerhoff, Stuart Eisendrath, J. Craig Nelson, Scott Mackin, Ian Gotlib, Stephen Chen, Eli Puterman, Blake Rawdin, Martin Picard, Laura Han.

2. Financial Support and Sponsorship: This work was supported by: NIMH grant R01 MH083784; The Tinberg Family; The O’Shaughnessy Foundation; UCSF Academic Senate.

Funding Disclosure: NIMH R01 MH083784

Footnotes

3.

The authors declare no conflicts of interest.

REFERENCES

  • 1.Viron MJ, Stern TA: The impact of serious mental illness on health and healthcare. Psychosomatics 2010, 51:458–465. [DOI] [PubMed] [Google Scholar]
  • 2.**.Lindqvist D, Epel ES, Mellon SH, et al. : Psychiatric disorders and leukocyte telomere length: Underlying mechanisms linking mental illness with cellular aging. Neurosci Biobehav Rev 2015, 55:333–364.-This article provides a comprehensive overview of telomere findings and mechanisms in psychiatric illnesses
  • 3.Muezzinler A, Zaineddin AK, Brenner H: A systematic review of leukocyte telomere length and age in adults. Ageing Res Rev 2013, 12:509–519. [DOI] [PubMed] [Google Scholar]
  • • 4.Wang Q, Zhan Y, Pedersen NL, et al. : Telomere Length and All-Cause Mortality: A Meta-analysis. Ageing Res Rev 2018, 48:11–20. [DOI] [PubMed] [Google Scholar]
  • 5.Darrow SM, Verhoeven JE, Revesz D, et al. : The Association Between Psychiatric Disorders and Telomere Length: A Meta-Analysis Involving 14,827 Persons. Psychosom Med 2016, 78:776–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sanders JL, Fitzpatrick AL, Boudreau RM, et al. : Leukocyte telomere length is associated with noninvasively measured age-related disease: The Cardiovascular Health Study. J Gerontol A Biol Sci Med Sci 2012, 67:409–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 7.Pepper GV, Bateson M, Nettle D: Telomeres as integrative markers of exposure to stress and adversity: a systematic review and meta-analysis. R Soc Open Sci 2018, 5:180744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 8.Lindqvist D, Simon NM, Wolkowitz OM: Is depression associated with accelerated aging? Mechanisms and implications. In Neurobiology of Depression: Road to Novel Therapeutics Edited by de Quevedo JL, Caravalho AF, Zarate CA: Academic Press; 2019:207–230. [Google Scholar]
  • • 9.Lin J, Sun J, Wang S, et al. : In vitro proinflammatory gene expression predicts in vivo telomere shortening: A preliminary study. Psychoneuroendocrinology 2018, 96:179–187. [DOI] [PubMed] [Google Scholar]
  • 10.Epel ES, Lin J, Wilhelm FH, et al. : Cell aging in relation to stress arousal and cardiovascular disease risk factors. Psychoneuroendocrinology 2006, 31:277–287. [DOI] [PubMed] [Google Scholar]
  • • 11.Athanasoulia-Kaspar AP, Auer MK, Stalla GK, Jakovcevski M: Shorter telomers associated with high doses of glucocorticoids: the link to increased mortality? Endocr Connect 2018. [DOI] [PMC free article] [PubMed]
  • 12.O’Donovan A, Pantell MS, Puterman E, et al. : Cumulative inflammatory load is associated with short leukocyte telomere length in the Health, Aging and Body Composition Study. PLoS One 2011, 6:e19687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 13.Manoliu A, Bosch OG, Brakowski J, et al. : The potential impact of biochemical mediators on telomere attrition in major depressive disorder and implications for future study designs: A narrative review. J Affect Disord 2018, 225:630–646. [DOI] [PubMed] [Google Scholar]
  • 14.*.Effros RB: Kleemeier Award Lecture 2008--the canary in the coal mine: telomeres and human healthspan. J Gerontol A Biol Sci Med Sci 2009, 64:511–515.-This is an excellent overview of inflammation- telomere interactions
  • 15.**.Blackburn EH, Epel ES, Lin J: Human telomere biology: A contributory and interactive factor in aging, disease risks, and protection. Science 2015, 350:1193–1198.-This is an excellent commentary on the role of telomere biology in stress and illness
  • 16.Shalev I, Moffitt TE, Braithwaite AW, et al. : Internalizing disorders and leukocyte telomere erosion: a prospective study of depression, generalized anxiety disorder and post-traumatic stress disorder. Mol Psychiatry 2014, 19:1163–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 17.Vance MC, Bui E, Hoeppner SS, et al. : Prospective association between major depressive disorder and leukocyte telomere length over two years. Psychoneuroendocrinology 2018, 90:157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hoen PW, de Jonge P, Na BY, et al. : Depression and leukocyte telomere length in patients with coronary heart disease: data from the Heart and Soul Study. Psychosom Med 2011, 73:541–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 19.Verhoeven JE, Revesz D, Picard M, et al. : Depression, telomeres and mitochondrial DNA: between- and within-person associations from a 10-year longitudinal study. Mol Psychiatry 2018, 23:850–857. [DOI] [PubMed] [Google Scholar]
  • 20.Verhoeven JE, van Oppen P, Revesz D, et al. : Depressive and Anxiety Disorders Showing Robust, but Non-Dynamic, 6-Year Longitudinal Association With Short Leukocyte Telomere Length. Am J Psychiatry 2016, 173:617–624. [DOI] [PubMed] [Google Scholar]
  • • 21.Chang SC, Crous-Bou M, Prescott J, et al. : Prospective association of depression and phobic anxiety with changes in telomere lengths over 11 years. Depress Anxiety 2018, 35:431–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.*.Gotlib IH, LeMoult J, Colich NL, et al. : Telomere length and cortisol reactivity in children of depressed mothers. Mol Psychiatry 2014.-An intriguing study showing shortened telomeres even before the onset of psychiatric illness
  • 23.Henje Blom E, Han LK, Connolly CG, et al. : Peripheral telomere length and hippocampal volume in adolescents with major depressive disorder. Transl Psychiatry 2015, 5:e676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Maurya PK, Rizzo LB, Xavier G, et al. : Shorter leukocyte telomere length in patients at ultra high risk for psychosis. Eur Neuropsychopharmacol 2017, 27:538–542. [DOI] [PubMed] [Google Scholar]
  • 25.Rackley S, Pao M, Seratti GF, et al. : Neuropsychiatric conditions among patients with dyskeratosis congenita: a link with telomere biology? Psychosomatics 2012, 53:230–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.*.Puterman E, Epel ES, Lin J, et al. : Multisystem resiliency moderates the major depression-telomere length association: findings from the Heart and Soul Study. Brain Behav Immun 2013, 33:65–73.Multi-system resiliency can buffer the effects of depression in telomere length
  • 27.Gotlib IH, LeMoult J, Colich NL, et al. : Telomere length and cortisol reactivity in children of depressed mothers. Mol Psychiatry 2015, 20:615–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 28.Powell TR, Dima D, Frangou S, Breen G: Telomere Length and Bipolar Disorder. Neuropsychopharmacology 2018, 43:445–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 29.Palmos AB, Breen G, Goodwin L, et al. : Genetic Risk for Psychiatric Disorders and Telomere Length. Front Genet 2018, 9:468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hjelmborg JB, Dalgard C, Moller S, et al. : The heritability of leucocyte telomere length dynamics. J Med Genet 2015, 52:297–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Codd V, Mangino M, van der Harst P, et al. : Common variants near TERC are associated with mean telomere length. Nat Genet 2010, 42:197–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Codd V, Nelson CP, Albrecht E, et al. : Identification of seven loci affecting mean telomere length and their association with disease. Nat Genet 2013, 45:422–427, 427e421–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hakobyan A, Nersisyan L, Arakelyan A: Quantitative trait association study for mean telomere length in the South Asian genomes. Bioinformatics 2016, 32:1697–1700. [DOI] [PubMed] [Google Scholar]
  • 34.Levy D, Neuhausen SL, Hunt SC, et al. : Genome-wide association identifies OBFC1 as a locus involved in human leukocyte telomere biology. Proc Natl Acad Sci U S A 2010, 107:9293–9298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mangino M, Hwang SJ, Spector TD, et al. : Genome-wide meta-analysis points to CTC1 and ZNF676 as genes regulating telomere homeostasis in humans. Hum Mol Genet 2012, 21:5385–5394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Pooley KA, Bojesen SE, Weischer M, et al. : A genome-wide association scan (GWAS) for mean telomere length within the COGS project: identified loci show little association with hormone-related cancer risk. Hum Mol Genet 2013, 22:5056–5064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Prescott J, Kraft P, Chasman DI, et al. : Genome-wide association study of relative telomere length. PLoS One 2011, 6:e19635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wei YB, Martinsson L, Liu JJ, et al. : hTERT genetic variation in depression. J Affect Disord 2016, 189:62–69. [DOI] [PubMed] [Google Scholar]
  • 39.Michalek JE, Kepa A, Vincent J, et al. : Genetic predisposition to advanced biological ageing increases risk for childhood-onset recurrent major depressive disorder in a large UK sample. J Affect Disord 2017, 213:207–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 40.Chang SC, Prescott J, De Vivo I, et al. : Polygenic risk score of shorter telomere length and risk of depression and anxiety in women. J Psychiatr Res 2018, 103:182–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 41.Verhoeven JE, Penninx B, Milaneschi Y: Unraveling the association between depression and telomere length using genomics. Psychoneuroendocrinology 2019, 102:121–127. [DOI] [PubMed] [Google Scholar]
  • 42.Wium-Andersen MK, Orsted DD, Rode L, et al. : Telomere length and depression: prospective cohort study and Mendelian randomisation study in 67 306 individuals. Br J Psychiatry 2017, 210:31–38. [DOI] [PubMed] [Google Scholar]
  • • 43.**.Declerck K, Vanden Berghe W: Back to the future: Epigenetic clock plasticity towards healthy aging. Mech Ageing Dev 2018, 174:18–29.–This is a very useful overview of epigentci clocks
  • 44.**.Horvath S: DNA methylation age of human tissues and cell types. Genome Biol 2013, 14:R115.-This is the landmark study that described DNA methylation patterns as “clocks” of human biological aging
  • 45.*.Chen BH, Marioni RE, Colicino E, et al. : DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging (Albany NY) 2016, 8:1844–1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.*.Hannum G, Guinney J, Zhao L, et al. : Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell 2013, 49:359–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • *47.Han LKM, Aghajani M, Clark SL, et al. : Epigenetic Aging in Major Depressive Disorder. Am J Psychiatry 2018, 175:774–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fries GR, Bauer IE, Scaini G, et al. : Accelerated epigenetic aging and mitochondrial DNA copy number in bipolar disorder. Transl Psychiatry 2017, 7:1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 49.Okazaki S, Otsuka I, Numata S, et al. : Epigenetic clock analysis of blood samples from Japanese schizophrenia patients. NPJ Schizophr 2019, 5:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.McKinney BC, Lin H, Ding Y, et al. : DNA methylation evidence against the accelerated aging hypothesis of schizophrenia. NPJ Schizophr 2017, 3:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 51.McKinney BC, Lin H, Ding Y, et al. : DNA methylation age is not accelerated in brain or blood of subjects with schizophrenia. Schizophr Res 2018, 196:39–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Voisey J, Lawford BR, Morris CP, et al. : Epigenetic analysis confirms no accelerated brain aging in schizophrenia. NPJ Schizophr 2017, 3:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.*.Zannas AS, Arloth J, Carrillo-Roa T, et al. : Lifetime stress accelerates epigenetic aging in an urban, African American cohort: relevance of glucocorticoid signaling. Genome Biol 2015, 16:266.-Interesting study of accelerated epigenetic aging with cumulative lifetime stress and the role of glucocorticoids
  • • 54.Verhoeven JE, Yang R, Wolkowitz OM, et al. : Epigenetic Age in Male Combat-Exposed War Veterans: Associations with Posttraumatic Stress Disorder Status. Mol Neuropsychiatry 2018, 4:90–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Boks MP, van Mierlo HC, Rutten BP, et al. : Longitudinal changes of telomere length and epigenetic age related to traumatic stress and post-traumatic stress disorder. Psychoneuroendocrinology 2015, 51:506–512. [DOI] [PubMed] [Google Scholar]
  • • 56.Mehta D, Bruenig D, Lawford B, et al. : Accelerated DNA methylation aging and increased resilience in veterans: The biological cost for soldiering on. Neurobiol Stress 2018, 8:112–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wolf EJ, Logue MW, Stoop TB, et al. : Accelerated DNA Methylation Age: Associations with PTSD and Mortality. Psychosom Med 2017. [DOI] [PMC free article] [PubMed]
  • • *58.Wolf EJ, Maniates H, Nugent N, et al. : Traumatic stress and accelerated DNA methylation age: A meta-analysis. Psychoneuroendocrinology 2018, 92:123–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 59.Wolf EJ, Logue MW, Morrison FG, et al. : Posttraumatic psychopathology and the pace of the epigenetic clock: a longitudinal investigation. Psychol Med 2019, 49:791–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Marioni RE, Harris SE, Shah S, et al. : The epigenetic clock and telomere length are independently associated with chronological age and mortality. Int J Epidemiol 2016, 45(2): 424–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zannas AS, Chrousos GP: Epigenetic programming by stress and glucocorticoids along the human lifespan. Mol Psychiatry 2017, 22:640–646. [DOI] [PubMed] [Google Scholar]
  • • 62.Ciccarone F, Tagliatesta S, Caiafa P, Zampieri M: DNA methylation dynamics in aging: how far are we from understanding the mechanisms? Mech Ageing Dev 2018, 174:3–17. [DOI] [PubMed] [Google Scholar]
  • • *63.Field AE, Robertson NA, Wang T, et al. : DNA Methylation Clocks in Aging: Categories, Causes, and Consequences. Mol Cell 2018, 71:882–895.-Very good review of the mechanisms of epigenetic aging
  • • 64.Irvin MR, Aslibekyan S, Do A, et al. : Metabolic and inflammatory biomarkers are associated with epigenetic aging acceleration estimates in the GOLDN study. Clin Epigenetics 2018, 10:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • *65.Morrison FG, Miller MW, Logue MW, et al. : DNA methylation correlates of PTSD: Recent findings and technical challenges. Prog Neuropsychopharmacol Biol Psychiatry 2019, 90:223–234.–Very good overview of methodological issues in testing epigenetic aging
  • • 66.Lu AT, Xue L, Salfati EL, et al. : GWAS of epigenetic aging rates in blood reveals a critical role for TERT. Nat Commun 2018, 9:387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • **67.Lu AT, Quach A, Wilson JG, et al. : DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY) 2019, 11:303–327.–Provocative, weel-done study of an epigenetic clock that purportedly predicts time-to-detah
  • 68.Levine ME, Lu AT, Bennett DA, Horvath S: Epigenetic age of the pre-frontal cortex is associated with neuritic plaques, amyloid load, and Alzheimer’s disease related cognitive functioning. Aging (Albany NY) 2015, 7:1198–1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rehkopf DH, Needham BL, Lin J, et al. : Leukocyte Telomere Length in Relation to 17 Biomarkers of Cardiovascular Disease Risk: A Cross-Sectional Study of US Adults. PLoS Med 2016, 13:e1002188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • **70.Picard M, McEwen BS: Psychological Stress and Mitochondria: A Systematic Review. Psychosom Med 2018, 80:141–153.–Excellent review of the role of stress in mitochondrial function
  • • 71.Theurey P, Pizzo P: The Aging Mitochondria. Genes 2018, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bersani FS, Morley C, Lindqvist D, et al. : Mitochondrial DNA copy number is reduced in male combat veterans with PTSD. Prog Neuropsychopharmacol Biol Psychiatry 2016, 64:10–17. [DOI] [PubMed] [Google Scholar]
  • • 73.Kumar P, Efstathopoulos P, Millischer V, et al. : Mitochondrial DNA copy number is associated with psychosis severity and anti-psychotic treatment. Sci Rep 2018, 8:12743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 74.Wang D, Li Z, Liu W, et al. : Differential mitochondrial DNA copy number in three mood states of bipolar disorder. BMC Psychiatry 2018, 18:149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.*.Cai N, Chang S, Li Y, et al. : Molecular signatures of major depression. Curr Biol 2015, 25:1146–1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 76.Lindqvist D, Wolkowitz OM, Picard M, et al. : Circulating cell-free mitochondrial DNA, but not leukocyte mitochondrial DNA copy number, is elevated in major depressive disorder. Neuropsychopharmacology 2018, 43:1557–1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 77.Tymofiyeva O, Henje Blom E, Ho TC, et al. : High levels of mitochondrial DNA are associated with adolescent brain structural hypoconnectivity and increased anxiety but not depression. J Affect Disord 2018, 232:283–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 78.Yamaki N, Otsuka I, Numata S, et al. : Mitochondrial DNA copy number of peripheral blood in bipolar disorder: The present study and a meta-analysis. Psychiatry Res 2018, 269:115–117. [DOI] [PubMed] [Google Scholar]
  • • 79.Kageyama Y, Kasahara T, Kato M, et al. : The relationship between circulating mitochondrial DNA and inflammatory cytokines in patients with major depression. J Affect Disord 2018, 233:15–20. [DOI] [PubMed] [Google Scholar]
  • • *80.Zhu Y, Liu X, Ding X, et al. : Telomere and its role in the aging pathways: telomere shortening, cell senescence and mitochondria dysfunction. Biogerontology 2019, 20:1–16. [DOI] [PubMed] [Google Scholar]
  • • 81.Zole E, Ranka R: Mitochondria, its DNA and telomeres in ageing and human population. Biogerontology 2018, 19:189–208. [DOI] [PubMed] [Google Scholar]
  • • 82.Herbers E, Kekalainen NJ, Hangas A, et al. : Tissue specific differences in mitochondrial DNA maintenance and expression. Mitochondrion 2019, 44:85–92. [DOI] [PubMed] [Google Scholar]
  • 83.Urata M, Koga-Wada Y, Kayamori Y, Kang D: Platelet contamination causes large variation as well as overestimation of mitochondrial DNA content of peripheral blood mononuclear cells. Ann Clin Biochem 2008, 45:513–514. [DOI] [PubMed] [Google Scholar]
  • 84.Clay Montier LL, Deng JJ, Bai Y: Number matters: control of mammalian mitochondrial DNA copy number. J Genet Genomics 2009, 36:125–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.**.Picard M, Juster RP, McEwen BS: Mitochondrial allostatic load puts the ‘gluc’ back in glucocorticoids. Nat Rev Endocrinol 2014, 10:303–310.-Excellent review of mitochondrial physiology and its relationship to stress, aging and illness
  • • 86.Guha M, Srinivasan S, Johnson FB, et al. : hnRNPA2 mediated acetylation reduces telomere length in response to mitochondrial dysfunction. PLoS One 2018, 13:e0206897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gonzales-Ebsen AC, Gregersen N, Olsen RK: Linking telomere loss and mitochondrial dysfunction in chronic disease. Front Biosci (Landmark Ed) 2017, 22:117–127. [DOI] [PubMed] [Google Scholar]
  • • 88.Osanai T, Tanaka M, Mikami K, et al. : Mitochondrial inhibitory factor protein 1 attenuates coupling factor 6-induced aging signal. J Cell Biochem 2018, 119:6194–6203. [DOI] [PubMed] [Google Scholar]
  • 89.Sanderson SL, Simon AK: In aged primary T cells, mitochondrial stress contributes to telomere attrition measured by a novel imaging flow cytometry assay. Aging Cell 2017, 16:1234–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kim H, Ham S, Jo M, et al. : CRISPR-Cas9 Mediated Telomere Removal Leads to Mitochondrial Stress and Protein Aggregation. Int J Mol Sci 2017, 18. [DOI] [PMC free article] [PubMed]
  • • 91.McCully KS: Review: Chemical Pathology of Homocysteine VI. Aging, Cellular Senescence, and Mitochondrial Dysfunction. Ann Clin Lab Sci 2018, 48:677–687. [PubMed] [Google Scholar]
  • 92.*.Sahin E, Depinho RA: Linking functional decline of telomeres, mitochondria and stem cells during ageing. Nature 2010, 464:520–528.-Excellent study of interactions between telomeres, telomerase and mitochondria
  • • 93.D’Aquila P, Montesanto A, De Rango F, et al. : Epigenetic signature: implications for mitochondrial quality control in human aging. Aging (Albany NY) 2019, 11:1240–1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.*.Riera CE, Merkwirth C, De Magalhaes Filho CD, Dillin A: Signaling Networks Determining Life Span. Annu Rev Biochem 2016, 85:35–64.–Excellent review of mitochondria and other signaling mechanisms is health and disease
  • • **95.Ferrucci L, Fabbri E: Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol 2018, 15:505–522.–Very good exposition of the concept of inflammaging and its health consequence
  • • *96.Squassina A, Pisanu C, Vanni R: Mood Disorders, Accelerated Aging, and Inflammation: Is the Link Hidden in Telomeres? Cells 2019, 8(1), 52.–Very good recent review
  • • 97.Misiak B, Frydecka D, Stanczykiewicz B, Samochowiec J: Editorial: Peripheral Markers of Immune Response in Major Psychiatric Disorders: Where Are We Now and Where Do We Want to Be? Front Psychiatry 2019, 10:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Franceschi C, Campisi J: Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci 2014, 69 Suppl 1:S4–9. [DOI] [PubMed] [Google Scholar]
  • 99.Kirkland JL, Tchkonia T: Cellular Senescence: A Translational Perspective. EBioMedicine 2017, 21:21–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Fougere B, Boulanger E, Nourhashemi F, et al. : Chronic Inflammation: Accelerator of Biological Aging. J Gerontol A Biol Sci Med Sci 2017, 72:1218–1225. [DOI] [PubMed] [Google Scholar]
  • 101.Xia S, Zhang X, Zheng S, et al. : An Update on Inflamm-Aging: Mechanisms, Prevention, and Treatment. J Immunol Res 2016, 2016:8426874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.van Deursen JM: The role of senescent cells in ageing. Nature 2014, 509:439–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • *103.Alpert A, Pickman Y, Leipold M, et al. : A clinically meaningful metric of immune age derived from high-dimensional longitudinal monitoring. Nat Med 2019, 25:487–495.–Intriguing new biological clock based on immune cell characteristics
  • • 104.Wolkowitz OM: Accelerated biological aging in serious mental disorders. World Psychiatry 2018, 17:144–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lupien SJ, Sasseville M, Francois N, et al. : The DSM5/RDoC debate on the future of mental health research: implication for studies on human stress and presentation of the signature bank. Stress 2017, 20:95–111. [DOI] [PubMed] [Google Scholar]
  • • 106.Culmsee C, Michels S, Scheu S, et al. : Mitochondria, Microglia, and the Immune System-How Are They Linked in Affective Disorders? Front Psychiatry 2018, 9:739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 107.Keenan CR, Allan RS: Epigenomic drivers of immune dysfunction in aging. Aging Cell 2019, 18:e12878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 108.Barnes RP, Fouquerel E, Opresko PL: The impact of oxidative DNA damage and stress on telomere homeostasis. Mech Ageing Dev 2019, 177:37–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 109.Kim Y, Vadodaria KC, Lenkei Z, et al. : Mitochondria, Metabolism, and Redox Mechanisms in Psychiatric Disorders. Antioxid Redox Signal 2019. [DOI] [PMC free article] [PubMed]
  • • 110.Vaiserman A: Developmental Tuning of Epigenetic Clock. Front Genet 2018, 9:584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • • 111.Wang J, Maxwell CA, Yu F: Biological Processes and Biomarkers Related to Frailty in Older Adults: A State-of-the-Science Literature Review. Biol Res Nurs 2019, 21:80–106. [DOI] [PubMed] [Google Scholar]
  • 112.Lopez-Otin C, Blasco MA, Partridge L, et al. : The hallmarks of aging. Cell 2013, 153:1194–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.*.Bersani FS, Lindqvist D, Mellon SH, et al. : Telomerase activation as a possible mechanism of action for psychopharmacological interventions. Drug Discov Today 2015, 20:1305–1309. [DOI] [PubMed] [Google Scholar]
  • 114.**.Verhoeven JE, Revesz D, Wolkowitz OM, Penninx BW: Cellular aging in depression: Permanent imprint or reversible process?: An overview of the current evidence, mechanistic pathways, and targets for interventions. Bioessays 2014, 36:968–978.-Very good overview of possible reversibility of cellular aging
  • • 115.Giraudeau M, Angelier F, Sepp T: Do Telomeres Influence Pace-of-Life-Strategies in Response to Environmental Conditions Over a Lifetime and Between Generations? Bioessays 2019, 41:e1800162. [DOI] [PubMed] [Google Scholar]
  • 116.**.Lin J, Epel E, Blackburn E: Telomeres and lifestyle factors: roles in cellular aging. Mutat Res 2012, 730:85–89.–Excellent account of lifestyle changes that may retard cellular aging
  • 117.*.Puterman E, Epel E: An intricate dance: Life experience, multisystem resiliency, and rate of telomere decline throughout the lifespan. Soc Personal Psychol Compass 2012, 6:807–825.-Psychological and social resiliency may offset cellular aging associated with depression
  • 118.*.Jaskelioff M, Muller FL, Paik JH, et al. : Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature 2011, 469:102–106.-Fascinating experiment showing reversibility of aging phenotypes by telomerase in animals
  • 119.**.Epel E: How “reversible” is telomeric aging? Cancer Prev Res (Phila) 2012, 5:1163–1168.-Readable and informative account of possible reversibility of telomere aging

RESOURCES