ArticlePDF AvailableLiterature Review

Aging Fits the Disease Criteria of the International Classification of Diseases

Authors:

Abstract

The disease criteria used by the World Health Organization (WHO) were applied to human biological aging in order to assess whether aging can be classified as a disease. These criteria were developed for the 11th revision of the International Classification of Diseases (ICD) and included disease diagnostics, mechanisms, course and outcomes, known interventions, and linkage to genetic and environmental factors. Results Biological aging can be diagnosed with frailty indices, functional, blood-based biomarkers. A number of major causal mechanisms of human aging involved in various organs have been described, such as inflammation, replicative cellular senescence, immune senescence, proteostasis failures, mitochondrial dysfunctions, fibrotic propensity, hormonal aging, body composition changes, etc. We identified a number of clinically proven interventions, as well as genetic and environmental factors of aging. Therefore, aging fits the ICD-11 criteria and can be considered a disease. This proposal was submitted to the ICD-11 Joint Task force, and this led to the inclusion of the extension code for “Ageing-related” (XT9T) into the “Causality” section of the ICD-11. These findings might lead to greater focus on biological aging in global health policy and might provide for more opportunities for the new therapy developers.
Contents lists available at ScienceDirect
Mechanisms of Ageing and Development
journal homepage: www.elsevier.com/locate/mechagedev
Aging Fits the Disease Criteria of the International Classication of Diseases
Daria Khaltourina
a,e
, Yuri Matveyev
b
, Aleksey Alekseev
c
, Franco Cortese
d
, Anca Ioviţă
e
a
Department of Risk Factor Prevention, Federal Research Institute for Health Organization and Informatics of Ministry of Health of the Russian Federation, Dobrolyubova
St. 11, Moscow, 127254, Russia
b
Research Lab, Moscow Regional Research and Clinical Institute, Schepkina St. 61/2 k.1, Moscow, 129110, Russia
c
Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory, GSP-1, Moscow, 119991, Russia
d
Biogerontology Research Foundation, Apt 2354 Chynoweth House, Trevissome Park, Truro, London, TR4 8UN, UK
e
International Longevity Alliance, 19 avenue Jean Jaurès, Sceaux, 92330, France
ABSTRACT
The disease criteria used by the World Health Organization (WHO) were applied to human biological aging in order to assess whether aging can be classied as a
disease. These criteria were developed for the 11th revision of the International Classication of Diseases (ICD) and included disease diagnostics, mechanisms, course
and outcomes, known interventions, and linkage to genetic and environmental factors.
Results: Biological aging can be diagnosed with frailty indices, functional, blood-based biomarkers. A number of major causal mechanisms of human aging involved
in various organs have been described, such as inammation, replicative cellular senescence, immune senescence, proteostasis failures, mitochondrial dysfunctions,
brotic propensity, hormonal aging, body composition changes, etc. We identied a number of clinically proven interventions, as well as genetic and environmental
factors of aging. Therefore, aging ts the ICD-11 criteria and can be considered a disease. Our proposal was submitted to the ICD-11 Joint Task force, and this led to
the inclusion of the extension code for Ageing-related(XT9T) into the Causalitysection of the ICD-11. This might lead to greater focus on biological aging in
global health policy and might provide for more opportunities for the new therapy developers.
1. Introduction
Aging (hereafter biological aging) can be dened as a complex in-
terrelated network of progressive functionally-deleterious phenotypic
deviations (with respect to the young adult phenotype) underlying
biological aging, leading to a decline in individualsadaptability, phy-
siological and mental function, as well as resilience.
There are discussions of whether aging should be classied as a
disease or not. A number researchers argued that aging should be
considered and treated as a disease (Bulterijs et al., 2015;de Grey and
Rae, 2007;Lakatta, 2015;Stambler, 2017;Zhavoronkov and Bhullar,
2015). The WHO developed a denition of disease for the purposes of
11th ICD revision:
A disease is a set of dysfunction(s) in any of the body systems
dened by: 1. Symptomatology: manifestations: known pattern of signs,
symptoms and related ndings 2. Aetiology: an underlying explanatory
mechanism 3. Course and outcome: a distinct pattern of development
over time 4. Treatment response: a known pattern of response to in-
terventions 5. Linkage to genetic factors: e.g., genotypes, patterns of
gene expression 6. Linkage to interacting environmental factors
(World Health Organization, 2011).
We undertook an eort to determine whether aging ts the ICD
denition of disease as per the current state of medical research. An
earlier version of this review was submitted to the WHO, which resulted
in the addition of the WHO ICD extension code "Ageing-Related" (XT9T)
for aging-related diseases in the Causalitysection of the ICD-11
(Calimport and Bentley, 2019).
2. Materials and methods
To obtain information on signs and symptoms of aging we screened
the most clinically relevant studies of aging biomarkers, including vast
amounts of research literature on frailty indices. The analysis of the
aetiology and underlying explanatory mechanisms of aging were per-
formed in several stages.
We initially produced a comprehensive list of aging-related patho-
logical processes, with some but not all of them being listed in ICD-10
as separate diseases, and others added from research literature
(Supplementary Table S1). We then screened medical research litera-
ture for reviews of the mechanisms of these aging-related pathological
processes and diseases in humans at the level of organs, tissues, cells,
signaling molecules, pathways, gene expression and interventions.
Animal research was reviewed as subsidiary when it covered important
knowledge gaps about certain aging-related cellular processes, mole-
cular pathways or epigenetic changes associated with aging and dis-
eases. We also reviewed pathogenetic processes of aging identied in
biogerontological research literature (de Grey and Rae, 2007;López-
Otín et al., 2013). Additionally, we documented clinical studies and
https://doi.org/10.1016/j.mad.2020.111230
Received 5 February 2019; Received in revised form 4 March 2020; Accepted 9 March 2020
E-mail address: khatourina@mednet.ru (D. Khaltourina).
Mechanisms of Ageing and Development 189 (2020) 111230
Available online 03 April 2020
0047-6374/ © 2020 Published by Elsevier B.V.
T
animal research on the eects of potential geroprotective interventions
on all-cause and cardiovascular mortality and other relevant clinical
endpoints in humans, as well as on lifespan of model organisms. We
documented the ndings on cellular and molecular mechanisms of
aging-related pathological processes and the benecial eects of ger-
oprotective interventions.
3. Results
3.1. Symptomatology and the course of disease
Large-scale cohort studies in humans and detailed investigations
into the biology of human aging provide individually validated bio-
markers and mechanisms, potentially leading to recommendations for
targeted interventions (Jia et al., 2017;Moeller et al., 2017).
3.1.1. Blood-based biomarkers of aging
A number of large studies identied sets of blood-based biomarkers
of aging which retrospectively predict morbidity and mortality in large
cohort studies and clinical trials (Bürkle et al., 2015;Horvath, 2013;
Moeller et al., 2017;Putin et al., 2016). These kinds of studies provide
tools for predicting clinically signicant aging-related outcomes, like
risks of morbidity, mortality, aging related diseases (Sebastiani et al.,
2017), cognitive or physical function decline or frailty.
Some proposed (Wagner et al., 2016) clinically signicant blood-
based biomarkers of aging include inammation agents, such as inter-
leukin-6 (IL-6) (Holly et al., 2013;Mitnitski et al., 2015;Sebastiani
et al., 2017;Soysal et al., 2016), TNF-a (Langmann et al., 2017;
Nascimento et al., 2018), C-reactive protein (CRP) (Sebastiani et al.,
2017;Soysal et al., 2016;Velissaris et al., 2017), and other in-
ammatory markers, glucose metabolism biomarkers such as glycated
hemoglobin (HbA1c) (Belsky et al., 2015) and plasma glucose
(Mamoshina et al., 2018;Wood et al., 2019;Zaslavsky et al., 2016),
adipokines (Mitnitski et al., 2015), homocysteine (Crimmins et al.,
2008;Mitnitski et al., 2015), thyroid hormones (Suzuki et al., 2012), N-
terminal prohormone of brain natriuretic peptide (NT-proBNP) (Draper
et al., 2018;Raymond, 2003), as well as troponin, albumin (Bouajila
et al., 2018) or albumin/creatinine ratio (Mitnitski et al., 2015;Putin
et al., 2016), lipid biomarkers such as low-density lipoprotein choles-
terol (LDL-C) (Carreira et al., 2012;Sebastiani et al., 2016), and tri-
glycerides (TGs) (Irvin et al., 2018;Rhee et al., 2019).
3.1.2. Function-based and anthropometric biomarkers
Clinically signicant physical function and anthropometric bio-
markers of aging include walking speed, standing from a chair, walking
balance, grip strength (Kim et al., 2018a), body mass index (BMI)
(Ismail et al., 2002;Muñoz et al., 2010), waist circumference, muscle
mass (Kim et al., 2016c) and others (Wagner et al., 2016).
3.1.3. Aging-related frailty and Frailty Indices
Fried et al. described frailty phenotype criteria including uninten-
tional weight loss, self-reported exhaustion, weakness (grip strength),
slow walking speed, and low physical activity (Fried et al., 2001).
Frailty Index was suggested (Mitnitski et al., 2001) as a proxy measure
of aging, and thus can be used (although crudely) as a metric that al-
lows researchers and clinicians to compare the rates of aging between
individuals and populations (Rockwood et al., 2005;Rockwood and
Mitnitski, 2007).
Prevalence of pre-frailty and frailty signicantly increases with age,
with pre-frailty prevalence reaching as high as 47% in old age, ac-
cording to some studies (Buckinx et al., 2015). Yet, frailty is related to
biological age stronger than to chronological age (Romero-Ortuno and
Kenny, 2012).
ICD-10 includes the R54 code for Senility, while ICD-10-CM codes
this condition as "Age-related physical debility". However, frailty has
become an established term after the introduction of the Rockwood
Frailty Scale in 2005 (Rockwood et al., 2005).
Frailty indices (FIs) are often used in epidemiological and clinical
studies to monitor health and to predict mortality and morbidity risks.
A number of similar FIs which are currently used (Jeong et al., 2013;
Rockwood et al., 2005) correlate better than calendar age with many
clinically signicant endpoints, including mortality (Ravindrarajah
et al., 2013), postoperative outcomes (Lin et al., 2016), sexual health
(Lee et al., 2012), cancer survival (Ommundsen et al., 2014), etc.
Frailty can be currently measured starting from the age as young as
15 (Rockwood et al., 2011). A number of lifestyle, nutraceutical and
pharmaceutical interventions have been proposed, developed and used
to prevent or reduce frailty levels (Cesari et al., 2015).
3.1.4. Combination of blood-based biomarkers with Frailty indices
Mitnitski et al. (Mitnitski et al., 2015) analyzed 40 biomarkers of
cellular aging, inammation, hematology and immunosenescence using
baseline data and 7-year mortality data from the Newcastle 85+ Study,
characterized the discriminatory ability of this biomarker-based frailty
index (FI-B) to predict mortality compared to individual biomarkers,
and then combined this FI-B with a clinical decits frailty index (FI-C)
to see whether the combined FI-B and FI-C index possessed greater
ability to predict mortality than the FI-B alone. The biomarkers which
were signicant predictors of mortality included blood-based in-
ammation biomarkers such as high sensitivity CRP, leptin, adipo-
nectin, homocysteine, albumin, hematological biomarkers (he-
moglobin, platelets, white blood cell count, neutrophils, lymphocytes),
biomarkers of immunosenescence, as well as transforming growth
factor β(TGF-β), and insulin-like growth factor-binding protein 1
(IGFBP1).
The researchers found that the FI-B has signicantly greater dis-
criminatory accuracy to predict mortality than any one biomarker or
biomarker subgroup alone, and that the combined FI-B and FI-C index
possessed the greatest overall ability to predict mortality.
3.1.5. Genetic biomarkers of aging
A number of genetic biomarkers of aging have been proposed, in-
cluding telomere length (Zhang et al., 2014b), T-cell DNA rearrange-
ment (Zubakov et al., 2010) and DNA methylation, as well as some
other epigenetic modications (Pal and Tyler, 2016).
Methylation patterns of certain CpG sites in DNA are highly corre-
lated with age. A number of epigenetic aging scores have been devel-
oped with high ability to predict mortality (Fransquet et al., 2019;Gao
et al., 2019) and calendar age including Horvaths epigenetic clock
(Horvath, 2013), epigenetic clock by Hannum et al. (Hannum et al.,
2013), GrimAge (Lu et al., 2019), and others (Horvath et al., 2018;
Weidner et al., 2014). Some epigenetic clocks measure accelerated
aging including DNAmPhenoAge acceleration (Levine et al., 2018) and
AgeAccelGrim (Lu et al., 2019). Data suggest that these epigenetic
aging clocks may be altered by lifestyle (Quach et al., 2017;Zhao et al.,
2019b) or, possibly, by some therapeutic interventions (Fahy et al.,
2019;Sae-Lee et al., 2018). Future clinical research will reveal how
relevant these indicators are in practice.
3.1.6. Indices of biological age
A large number of indices of biological age have been proposed.
Some biological age formulas measure biological aging per se with a
number of biomarkers (Jia et al., 2017), while a number of research
groups use Klemera and Doubals method in such indices, which adds
age acceleration (deceleration) to calendar age (Klemera and Doubal,
2006). Other groups used indicators of pathologies (Hochschild, 1989)
or articial intelligence methods to create indices of biological aging
(Putin et al., 2016;Pyrkov et al., 2018;Rahman and Adjeroh, 2019).
In sum, there is a growing number of diagnostic tools which can be
used to assess the rate of biological aging, to develop new medicines
and to monitor eectiveness of interventions. Research on FIs reached
considerable degree of consensus, while the research on blood-born,
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
2
epigenetic and other biomarkers is gaining momentum. Therefore,
aging diagnostics is already available and is developing further.
3.2. Aetiology: an underlying explanatory mechanism
We performed a search in the medical research literature and
identied a number of interrelated pathological processes which are
known to take part in the pathological aging-related processes and
changes of various systems and structures of the human body listed in
Table S1.
3.2.1. Chronic low-grade inammation
Aging-related chronic low-grade inammation ("inamm-aging") is
described by the increase in inammatory markers and agents in aging,
in the absence of overt infection (asepticinammation).
There is overwhelming epidemiological evidence that the state of
mild inammation, revealed via elevated levels of inammatory bio-
markers such as CRP, IL-6 (Mitnitski et al., 2015;Soysal et al., 2016),
and potentially a few other inammation biomarkers, is predictive of
many aging phenotypes, such as changes in body composition, energy
production and utilization, metabolic homeostasis, immune senescence,
neuronal health and frailty (Franceschi and Campisi, 2014).
Circulating proinammatory molecules are strong predictors of age-
related morbidity and mortality (Barron et al., 2015;Li et al., 2017b).
There is mounting clinical evidence that local production of in-
ammatory cytokines can drive phenotypes and pathologies associated
with aging, including atherosclerosis (Lorenzatti and Retzla, 2016),
diabetes (Park et al., 2014), pulmonary diseases (Franceschi and
Campisi, 2014;Murray and Chotirmall, 2015), and Alzheimer's disease
(AD) (Franceschi and Campisi, 2014).
Neuroinammation is the major route of nervous system aging and
degenerative disease development (Yin et al., 2016).
Aging-related increase in the levels of pro-inammatory cytokines is
caused by replicative cellular senescence (Coppé et al., 2010), increased
body fat share, mitochondrial damage, damaged macromolecules (Shi
et al., 2015) and cells (self-debris) that accumulate with age due to
increased production and/or inadequate elimination (Franceschi and
Campisi, 2014) and microbiota changes (Nagpal et al., 2018).
Exercise (Monteiro-Junior et al., 2018) and the decreased BMI leads
to decrease in serum inammatory markers (Fedewa et al., 2017). Both
physical exercise (Naci and Ioannidis, 2013) and intentional weight loss
in people with preexisting obesity (Kritchevsky et al., 2015) are asso-
ciated with reduced all-cause mortality.
Healthy dietary patterns are also associated with lower in-
ammatory markers (Soltani et al., 2017). The consumption of olive oil
(Schwingshackl et al., 2015) as well as nuts (Neale et al., 2017) de-
creases inammation markers and reduces the risks of cancer incidence
(Pelucchi et al., 2011), cardiovascular and all-cause mortality (Chen
et al., 2017b;Schwingshackl and Homann, 2014). Coee consumption
also reduces serum concentration of CRP (Zhang and Zhang, 2018), and
may reduce cancer incidence (Poole et al., 2017).
Vitamin D supplementation decreases CRP level (Mirhosseini et al.,
2018), and it may also reduce all-cause mortality in adults (Chowdhury
et al., 2014) and cancer risks (Grant, 2018), perhaps due to high rates of
vitamin D deciency in the modern societies. There is evidence that the
use of aspirin, an anti-inammatory drug, is associated with modestly
reduced all-cause, cardiovascular mortality (in non-hypertensive sub-
jects) and cancer prevalence, as well as mortality in older adults
(Sutclie et al., 2013;Whitlock et al., 2015).
Therefore, all the above-mentioned interventions can be viewed as
both anti-inammatory and geroprotective (capable of addressing pa-
thological aging processes and extending lifespan in humans).
Glucosamine and chondroitin use reduces CRP in healthy subjects
(Navarro et al., 2015), and may be associated with reduction in all-
cause mortality, mortality from cancer and respiratory diseases, ac-
cording to the observational studies (Bell et al., 2012).
Other potential anti-inammatory interventions with some systemic
health benets include acipimox (Liang et al., 2013), quercetin
(Mohammadi-Sartang et al., 2017;Sahebkar, 2017;Serban et al., 2016),
coenzyme Q10 (CoQ10) (Zhai et al., 2017), ginger (Mazidi et al., 2016),
resveratrol (Guo et al., 2017), magnesium (Simental-Mendia et al.,
2017), and arnesin/carnosine (Hisatsune et al., 2016;Menon et al.,
2018), melatonin (Akbari et al., 2018), curcumin (Tabrizi et al., 2018b),
liraglutide (Kahal et al., 2014), telmisartan (Takagi et al., 2013), sil-
denal(Santi et al., 2015), pentoxifylline (Pollice et al., 2001), as well
as probiotic (Mazidi et al., 2017), prebiotic and symbiotic supplements
(McLoughlin et al., 2017).
3.2.2. Replicative cellular senescence
Cellular senescence refers to the durable arrest of cell proliferation
that occurs when cells experience certain types of alterations, including
telomere shortening and activation of damage sensing pathways, such
as p38 mitogen-activated protein kinases (p38
MAPK
) and nuclear factor
kappa-light-chain-enhancer of activated B cells (NF-κB) (Campisi, 2013;
He and Sharpless, 2017). The majority of senescent cells can be de-
tected via beta-galactosidase assay, estimation of Lamin B1 loss and
markers of senescence-associated secretory phenotype (SASP).
Both telomere shortening in various tissues (Ishikawa et al., 2016)
and the accumulation of post-mitotic senescent cells increase with age
(Belsky et al., 2015;Liu et al., 2009;Sørensen et al., 2016).
Telomere length (TL) correlates negatively with the risks of cardi-
ovascular (Akasheva et al., 2015;Dudinskaya et al., 2015;Haycock
et al., 2014;Strazhesko et al., 2015), neurodegenerative (Forero et al.,
2016), lung (Calhoun et al., 2016) diseases, and mortality in people
aged 60 and over (Cawthon et al., 2003;Sørensen et al., 2016), as well
as in cancer survivors (Xu et al., 2016).
Telomere shortening is a major pathway of cellular senescence.
When telomeres reach critical shortness, the cell enters either senes-
cence, normally through the induction of cyclin-dependent kinase in-
hibitor 2A (p16
INK4a
), or apoptosis. The intracellular enzyme telo-
merase can promote telomere lengthening, preventing the age-related
shortening that comes with cell division.
Additionally, cellular senescence can be induced by stressors like
mitochondrial or DNA damage, oxidative stress (Reichert and Stier,
2017), as well as the use of cytotoxic/genotoxic therapies.
The causal mechanisms for the associations between age-related
increase in cellular senescence and the disease pathogenesis include
pro-inammatory and pro-brotic SASP of senescent cells, the loss of
proliferation-competent cells, and the reduction of the total number of
functional tissue cells.
SASP has been shown to drive local and systemic inammation
accompanied by macrophage and lymphocyte inltration, apoptosis
and brosis. Recent evidence in broblasts and epithelial cells has
shown that cellular senescence is accompanied by a striking increase in
the secretion of 4080 factors that participate in intracellular signaling.
SASP proteins are generally induced at the level of mRNA and include a
wide range of growth factors, proteases, chemokines and cytokines.
Proteins that are known to stimulate inammation, including IL-6, IL-8,
IL-1, granulocyte macrophage colony-stimulating factor (GM-CSF),
growth regulated oncogene (GRO)-α, monocyte chemotactic proteins
(MCPs) MCP-2, MCP-3, matrix metalloproteinases (MMPs) MMP-1,
MMP-3, and many of the insulin-like growth factor-binding proteins
(IGFBPs), are among the most robustly induced and secreted of these
factors (Freund et al., 2010).
While cellular senescence may play a protective role in some con-
ditions like wound healing, cancer or brosis, the burden of senescent
cells seems to be net-detrimental for aging organisms. Replicative arrest
is oncoprotective, however, on the other side, SASP induction by the
senescent cells might promote cancer. Also, senescent cells can com-
promise tissue cancer resistance capacity. These ndings open oppor-
tunities for new therapeutic approaches to the treatment of age-related
diseases (Hodes et al., 2016).
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
3
There are numerous examples of involvement of senescent cells in
the pathogenesis of the diseases of aging, including CVDs (Alique et al.,
2018;Krouwer et al., 2012;Wang and Bennett, 2012), AD (Bhat et al.,
2012;Ojo et al., 2015), glaucoma (Liton et al., 2005), cataract (Zhou
et al., 2015), and osteoarthritis (McCulloch et al., 2017).
The interventions which prevent or reduce the harmful eects of
cellular senescence are still being explored.
Physical activity (Mundstock et al., 2015) and healthy dietary pat-
terns, such as Mediterranean diet, as well as fruit and vegetable con-
sumption might be related to longer telomere length (TL), while con-
sumption of certain food categories including processed meat, cereals
and sugar-sweetened beverages, as well as excessive red meat con-
sumption may be associated with shorter TL (Rae et al., 2017).
Also, metformin use might be associated with larger telomere length
in prediabetic subjects (de Kreutzenberg et al., 2015).
Statins modulate telomerase activity and reduce telomere erosion
(Boccardi et al., 2013;Paradisi et al., 2012), while atorvastatin acti-
vates telomerase in patients free of atherosclerosis (Strazhesko et al.,
2016).
Some other potential pharmacological interventions against cellular
senescence have been identied, but their eects and toxicity still need
to be studied. For example, quercetin is known to promote apoptosis in
senescent cells (Khan et al., 2016).
Cycloastragenol (TA-65, Astragalus propinquus extract) is pur-
ported to have telomerase activation activity. Astragalus may be ben-
ecial for patients with non-small-cell lung (He et al., 2013) and he-
patocellular cancers (Tian et al., 2016;Wu et al., 2009), and early
macular degeneration (Dow and Harley, 2016).
Lubricant eye drops with N-acetylcarnosine, a carnosine prodrug, at
physiological concentration might remarkably reduce the rate of telo-
mere shortening in the lens and improve visual functions in older
people (Babizhayev and Yegorov, 2016). At the same time, carnosine
supplementation may reduce oxidative stress (Regazzoni et al., 2016),
while anserin and carnosine supplements might improve cognitive
abilities and physical functioning in this group (Hisatsune et al., 2016;
Szcześniak et al., 2014).
Olive phenols might prevent cellular senescence (Menicacci et al.,
2017), but further research is needed.
3.2.3. Proteostasis failures
Protein homeostasis (proteostasis) is a network of biological path-
ways responsible for protein folding, tracking, metabolism and de-
gradation within and outside cells, which is essential for health main-
tenance. Pathological processes of aging impose signicant chronic
stress on proteostatic systems, which induces various subsequent cas-
cades of pathogenesis.
Aggregations of misfolded proteins in proteinaceous inclusions are
common to many age-related diseases, including cardiovascular,
kidney, liver, eye diseases, osteoporosis (Yamagishi et al., 2015), dia-
betes (Nenna et al., 2015), as well as neurodegenerative diseases, such
as AD and Parkinson's disease (PD) (Chiti and Dobson, 2017;Iannuzzi
et al., 2014).
Misfolded protein aggregation is found not only in patients with
certain diseases, for example, signicant amyloid deposits are found in
20 to 30% of the brains of healthy older adults (Rodrigue et al., 2009).
Damaged macromolecules accumulate with age due to increased
production and/or inadequate elimination of certain proteins which
promotes pathological processes, both specic, including cognitive
decline and type 2 diabetes (T2D), and non-specic such as in-
ammation.
There are dierent mechanisms of misfolded protein toxicity.
Smaller oligomers are the major pathogenic source of the neuropathic
diseases of the central nervous system, as they interact with various
cellular structures including phospholipid bilayers of the cell membrane
and a number of cell receptors. At the same time, pathogenic species in
non-neuropathic systemic or localized amyloidoses include both
extracellular amyloid deposits aecting organ integrity and protein
oligomers that are produced during the process of their formation or are
released by mature deposits, causing direct cellular damage (Chiti and
Dobson, 2017).
Some misfolded proteins, including amyloid-beta (Aβ)(Lucia
Pagani, 2011), amylin (Lim et al., 2010), α-synuclein (Chiti and
Dobson, 2017), advanced glycation end products (AGEs) (Neviere et al.,
2016), advanced oxidation protein products (AOPPs) (Ye et al., 2017)
detrimentally aect mitochondrial function, induce oxidative stress and
apoptosis.
Healthy low levels of Aβpeptides might be needed for cGMP-in-
duced long-term potentiation and memory (Palmeri et al., 2017).
However, excess of Aβoligomers is a major contributor to AD, as they
interact with the phospholipid bilayers of the cell membrane, N-Methyl-
D-aspartic acid (NMDA) and AMPA receptors, the metabotropic gluta-
mate receptor 5, the insulin receptor, the nicotinic acetylcholine re-
ceptor α7-nAcChR, cellular prion protein (PrP
C
), and other cellular
components and membrane receptors leading to cognitive decline and
AD development (Benilova et al., 2012;Chiti and Dobson, 2017).
Misfolded tau protein is another major contributor of AD (Chiti and
Dobson, 2017).
Similarly, glycation of α-synuclein is a factor involved in the protein
aggregation in the formation of Lewy bodies and in PD development.
Age-related amylin accumulation induces inammatory response in
pancreatic islets and apoptosis in insulin-producing beta cells, which is
strongly implicated in the progression of T2D (Singh et al., 2015). They
may also play a role in the pathogenesis of cardiac (Despa et al., 2012)
and cognitive dysfunctions (Ly and Despa, 2015;Qiu and Zhu, 2014).
AGEs are a diverse group of macromolecules formed through sugars
covalent bonding to proteins, lipids, or nucleic acids as result of non-
enzymatic reaction, and they accumulate in organisms during aging.
There is a growing body of evidence that AGEs induce oxidative
stress generation through their receptor (RAGE) and as a result stimu-
late inammatory, thrombotic, brotic and proliferative responses in a
variety of cells and tissues. AGEs and RAGE are involved in the devel-
opment of diabetes and aging-associated disorders such as cardiovas-
cular complications (Neviere et al., 2016), cancer, AD, osteoporosis
(Yamagishi et al., 2015), chronic kidney disease (CKD) (Nakamura
et al., 2010) nonalcoholic steatohepatitis (NASH) (Takeuchi et al.,
2017).
Some misfolded proteins damage-associated biomolecular sig-
natures initiate innate immune responses by being recognized as
dangersignals by a network of sensors (including but not limited to
the Nlrp3 inammasome) (Shi et al., 2015).
The burden of misfolded proteins increases in aging via dierent
mechanisms.
Proteins and other signaling molecules undergo various forms of
enzymatic and non-enzymatic post-translational modications. Many of
these proteins are vital for the normal organism functioning, but
changes in the frequency of some of these modications can lead to
dysfunction. Examples of protein post-translational modications that
have been suggested to have causal links to aging, include glycation,
various forms of oxidative damage, nitration, enzymatic crosslinks,
deamidation, racemization, isomerization, and carbamylation.
Additionally, changes in glycosylation appear to act as clinical bio-
markers of aging in humans (Ding et al., 2011;Vanhooren et al., 2010).
Some of the health problems imposed by protein metabolism dis-
balances are caused by developmental processes and can be referred to
as programmed events. The most prominent example is elastic tissue
degradation, which leads to blood vessel stiening, as well as aging of
lungs, skin, ligaments, etc.
The major part of elastic ber formation takes place during prenatal
and early neonatal periods and is suppressed in adults by post-tran-
scriptional factors (Mora Huertas et al., 2016).
Additionally, elastin-derived peptides (EDPs) derived from elastic
tissue degradation, are involved in vascular, lung and renal diseases
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
4
(Duca et al., 2016;Smith et al., 2012). Elastolysis is mainly triggered by
elastases from inammatory cells that secrete neutrophil elastase,
MMPs (especially MMP-2), and cathepsins. Higher serum levels of
MMP-2, cathepsins and EDPs were independently associated with
baseline aortic pulse wave velocity, and MMP-2 and EDP levels in-
dependently predicted all-cause mortality in a cohort study of people
with predialysis CKD (Smith et al., 2012). The downstream signaling of
EDPs involves spliced-galactosidase (s-Gal), NF-κB, Sialidase 1 (Neu-1),
ERC, M3 ganglioside, lactosylceramide, etc. (Duca et al., 2016).
Inadequate protein clearance is a mechanism implicated in many
pathological processes of aging. For example, decreased or inadequate
clearance of Aβleads to the accumulation of Aβoligomers and larger
aggregate formation (Kundra et al., 2017). Aging-related increase in
oxidative stress and decrease in mitochondrial function reduce the ef-
fectiveness of the proteostasis networks (Chiti and Dobson, 2017).
Many proteostatic processes and systems are impaired or over-
loaded during aging. Thus, the function of the chaperome, the ensemble
of chaperones and co-chaperones that interact in a complex network of
molecular folding machines to regulate proteome function, is drama-
tically repressed in human aging brains and in the brains of patients
with neurodegenerative diseases (Brehme et al., 2014).
Autophagy and proteasome activities have both been found to de-
crease with aging (Chiti and Dobson, 2017;Morimoto and Cuervo,
2014), but to a lesser extent in healthy centenarians (Emanuele et al.,
2014). Defective autophagy is assumed to play a role in some aging-
related conditions, like osteoarthritis (Li et al., 2016;Nakamura et al.,
2010), AD, PD (Kaushik and Cuervo, 2015), and cancer (Xie and Zhou,
2018).
Interventions to reduce the burden of misfolded proteins vary de-
pending on the protein species. Some of them are being explored yet
(Bartolome et al., 2018;Zhou et al., 2016).
Proper dietary (Hanson et al., 2013) and sleep regimes (Ooms et al.,
2014) might be helpful to reduce Aβlevels in the nervous system in at
least some demographic groups.
There is some evidence that 17β-estradiol administration reduces
plasma Aβ40 in HRT-naïve postmenopausal women (Baker et al.,
2003), although safety and eectiveness of various regimes of hormone
replacement therapy (HRT) against dementias for women is being
questioned (Marjoribanks et al., 2017).
Some innovative therapies like plasma exchange with 5% albumin
are promising against accumulation of Aβpeptides (Boada et al., 2017;
Cuberas-Borrós et al., 2018).
Anatural product dihydroergocristine can reduce the burden of Aβ
(Lei et al., 2015). Dihydroergocristine use may have been associated
with better survival in dementia patients in one cohort study (Wu et al.,
2015a).
Simvastatin use leads to decreased levels of misfolded p-tau181
protein in cerebrospinal uids (Li et al., 2017a;Riekse et al., 2006).
Statin use may be associated with lower risks of cognitive decline
(Smith et al., 2017) and dementia (Chou et al., 2014;Geifman et al.,
2017).
Many interventions which reduce the burden of AGEs are known to
have benecial health eects in older adults, including dietary mod-
ications (Uribarri et al., 2003), anti-diabetic drugs (sodium/glucose
cotransporter 2 (SGLT2) inhibitors (Bekki et al., 2018)), some of an-
giotensin II receptor blockers (ARBs) (Honda et al., 2012;Rabbani
et al., 2012), some statins (Cuccurullo et al., 2006;Fukushima et al.,
2013;Kimura et al., 2010), or anti-oxidants such as carnosine
(Babizhayev et al., 2012), quercetin (Byun et al., 2017;Nenna et al.,
2015), and curcumin with Boswellia serrata (Chilelli et al., 2016).
3.2.4. Immune senescence
Aging is a risk factor for the immune systems dysfunction (immune
senescence), which leads to signicantly increased vulnerability to in-
fections, and poorer response to newly encountered antigens, as well as
weaker responses to vaccines and immunotherapies for cancer and
other diseases common in the older demographics (Pera et al., 2015).
A major causal mechanism of immune senescence is the decline in
the number of naïve T-cells (Vescovini et al., 2014) due to thymic in-
volution, T-cell cellular senescence and impaired homeostatic pro-
liferation (Bellon and Nicot, 2017). The loss of naïve T-cell population
correlates with frailty (Johnstone et al., 2017), mortality (Mitnitski
et al., 2015) and well as with the risks of certain non-communicable
diseases (Moro-García et al., 2015).
Aging-related metabolic changes, including hormonal changes (Pera
et al., 2015), low-grade inammation, increased exposure of T-cells to
pro-inammatory molecules, DNA damage and expansion of dysfunc-
tional T-cells clones are primary causal factors implicated in the pa-
thogenesis and progression of immune senescence.
At the same time, persistent viral load through lifespan and that of
cytomegalovirus (CMV), in particular is the major promoter of immune
senescence (Bellon and Nicot, 2017). CMV is highly prevalent world-
wide and persists in its host for life. In synergy with aging, it promotes
dierentiation of CD8+ T-cells, and the acquisition of a low CD4:CD8
T-cell ratio, which is a marker of aging-related or a virus-mediated
depletion of CD8+ naïve T-cell pools, and which correlates in-
dependently with frailty, morbidity and mortality in older people
(Johnstone et al., 2017;Mitnitski et al., 2015;Weltevrede et al., 2016;
Wikby et al., 1998). This suggests that the loss of immune-competent T-
cells in combination with CMV persistence are among causal mechan-
isms of aging and age-related morbidity and mortality.
Some potential geroprotective interventions are known to improve
T-cell immunity. Regular exercise increases naïve T-cell count (Duggal
et al., 2018), improves immune function and results in lower incidence
of disease (Arem et al., 2015;World Health Organization, 2010). Fre-
quent acute (but not extreme) physical exercise may increase relative
CD8+ T-cell telomere length, and allow naïve T-cells to occupy "va-
cant" immune space and increase the naïve T-cell repertoire (Simpson
et al., 2010). Vaccine response in adults can be improved with pro-
biotics (Lei et al., 2017) or mammalian target of rapamycin (mTOR)
inhibitors (Mannick et al., 2014). Antiviral therapies which are eective
against CMV and some other viruses and can reverse the development
of immune senescence in mice (Beswick et al., 2013), and their pro-
phylactic use might reduce mortality among renal transplant (Reddy
et al., 2003) and allogeneic stem cell transplant (Marty et al., 2017)
patients. Long-term safety and eectiveness of these interventions in
terms of preventing immunosenescence needs to be explored.
3.2.5. Increased brotic propensity
Aging is associated with signicantly increased risks of chronic -
broproliferative conditions, including hepatic, renal, heart, vascular,
and pancreatic brosis (Rosenbloom et al., 2017), idiopathic lung -
brosis (IPF), chronic obstructive lung disease (COPD), infertility (Briley
et al., 2016), erectile dysfunction (El-Sakka, 2011), (Karsdal et al.,
2014), as well as eye diseases with brotic components (glaucoma,
secondary cataract (Rosenbloom et al., 2017), retinopathies (Roy et al.,
2016), and dry eye disease).
Fibrosis develops as a reaction to damage, such as mechanical da-
mage, toxins, inammation, oxidative stress, re-
ninangiotensinaldosterone system (RAAS) overactivation, hypoxia,
which all may lead to pathological accumulation of extracellular matrix
(ECM) protein. Formation of excess brous connective tissue in an
organ or tissue occurs as part of a reparative or reactive process (e.g.
wound healing). When a critical organ such as the heart, liver, kidney
or pancreas is aected by brosis it can lead to the loss of its integrity
and function, which can result in further cascades of systemic damage.
Also, reduced blue light transmission due to aging-related brotic eye
diseases may impair the photoentrainment of circadian rhythm leading
to sleep disorders (Ayaki et al., 2016), and, possibly, higher levels of
systemic inammation (Kessel et al., 2011).
The TGF-βsuperfamily is the most important ECM regulator, which
plays important roles in embryogenesis and development, with TGF-β1
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
5
being the major pro-brotic protein (Hu et al., 2018). TGF-β1 signaling
increases with age (Parker et al., 2017;Wang et al., 2006), and it can be
exacerbated by diabetes (Meng et al., 2016;Mou et al., 2016;Qiao
et al., 2017). Insulin resistance (IR) and obesity-derived inammation
may also lead to meta-brosis(excessive ECM formation) (Lark and
Wasserman, 2017).
Dysregulated signaling between certain molecules and pathways,
including TGF-β, in particular TGF-β1, SMAD, SNAI1, CTGF (con-
nective tissue growth factor) (Lipson et al., 2012), vascular endothelial
growth factor (VEGF) (Chaudhary et al., 2007), epidermal growth
factor receptor (EGFR), galectin-3 and lysyl oxidase homolog 2 (LOXL2)
as well as MTORC1 and MTORC2 via PI3K/AKT and Wnt pathways
(Meng et al., 2016), which can be stimulated by reactive oxygen species
(ROS) induced by reduced form of nicotinamide adenine dinucleotide
phosphate (NADPH) oxidases (especially Nox4), inammatory cyto-
kines, such as IL-6, IL-1a, IL-13, IL-4, tumor necrosis factor (TNF-α)
(Lark and Wasserman, 2017), hypoxia (hypoxia-inducible factor 1-α,
HIF-1-α) and leading ultimately to endothelial-mesenchymal transition
and brosis formation (Li et al., 2017c). Activated broblasts, pericytes
and other cells increase production of collagen, bronectin, α-smooth
muscle actin (α-SMA) and other ECM proteins. This leads to the pro-
duction and accumulation of sub-pools of cross-linked proteins that can
be termed the end productsof brosis. Hypoxia, one of the systemic
factors of aging, also acts to increase SNAI1 level mediated by HIF-1-α.
Mechanisms of tissue remodeling and inammation resolution leading
to clearance of brotic matrix by extracellular proteolysis and/or en-
docytosis are inadequate in brosis (Piera-Velazquez et al., 2016).
Within the course of brosis formation, proteases (such as MMP-9,
MMP-12, MMP-13), secreted as a part of the damage response, degrade
the existing ECM components and release various protein fragments
(neo-epitopes) into circulation (Giannandrea and Parks, 2014).
Fibrosis research is still in development. A number of interventions
with known anti-aging eects in certain groups of patients are shown to
inhibit liver brosis progression in humans, including healthy diet
(Perumpail et al., 2017), coee consumption (Liu et al., 2015), statins
(Kim et al., 2017), aspirin (Poujol-Robert et al., 2014;Shen et al.,
2014), RAAS inhibitors (Kim et al., 2016b), and, possibly, liraglutide
(Kahal et al., 2014). In fact, RAAS targeting drugs, such as ACE in-
hibitors and ARBs prevent and sometimes even regress myocardial,
renal, hepatic, and muscle brosis and alleviate COPD symptoms (Garg
et al., 2015;Shrikrishna et al., 2012).
N-Acetylcysteine (NAC), an antioxidant drug, improves symptoms
of COPD (Firuzi et al., 2011), and it could be eective in other brotic
conditions, including renal diseases (Ahmadi et al., 2017), liver failure
(Chughlay et al., 2016), myocardial infarctions (Talasaz et al., 2014).
Dasatinib plus quercetin combination may alleviate idiopathic pul-
monary brosis (Justice et al., 2019).
CoQ10 and selenium supplementation may lead to less brosis in
the older people, which may explain lower cardiovascular and all-cause
mortality in one study (Alehagen et al., 2018;Lehagen et al., 2018).
Additionally, cGMP-specic phosphodiesterase type 5 (PDE5) in-
hibitors, the NO-inducing drugs that address erectile dysfunction, seem
not only to prevent and reverse penile brosis (El-Sakka, 2011), but
also to reduce mortality in men with CVDs including myocardial in-
farctions (Andersson et al., 2017) and diabetes (Hackett et al., 2017).
Pentoxifylline, a non-selective phosphodiesterase (PDE) inhibitors may
be eective against miscellaneous brotic conditions (Wen et al., 2017)
and preserve kidney function in CKD (Chen et al., 2017c).
A number of interventions reduce arterial stiness, including phy-
sical exercise (Zhang et al., 2018b) and RAAS blockers and metformin
(Wu et al., 2015a).
3.2.6. Aging-related mitochondrial dysfunctions
Mitochondria are organelles in charge of energy (adenosine tri-
phosphate, ATP) production, cellular respiration, storage of calcium
ions (Ca
2+
), regulation of cellular apoptositis, etc.
Mitochondrial respiratory chain dysfunction is a hallmark of aging
and manifests in decreased ATP and nicotinamide adenine dinucleotide
+ (NAD+) production, as well as excessive oxidative stress. It is in-
volved in major pathogenetic processes of aging, including inamma-
tion (Picca et al., 2018), cellular senescence (Reichert and Stier, 2017;
Schuliga et al., 2018), IR (Jin and Diano, 2018;Tardito, 1990), im-
paired proteostasis (Chiti and Dobson, 2017;Kaushik and Cuervo,
2015), decreased stem cell functionality (Zhang et al., 2018a), muscle
loss (Coen et al., 2013;Doria et al., 2012), and neurodegeneration
(Brooks et al., 2007;Mosconi et al., 2006).
Aging-related mitochondrial dysfunctions aect multiple body sys-
tems, while energy-intensive organs like muscles (Coen et al., 2013;
Doria et al., 2012) or the nervous system are particularly vulnerable.
For example, AD is also accompanied by decreased mitochondrial
bioenergetics (Brooks et al., 2007), which can occur as early as a decade
before the diagnosis (Mosconi et al., 2006).
The hallmarks of aging-related mitochondrial dysfunctions include
mitochondria loss in aging, decreased ATP and NAD+ production,
impaired mitochondrial membrane function, swollen mitochondria,
damaged cristae, decreased mitochondrial DNA copy number, and ex-
cessive burden of ROS (Giuseppe and Ross, 2016). Decline in mi-
tochondrial biogenesis, cellular NAD+, ATP levels, decreased NAD
+/NADH ratio (Ying, 2008;Zhu et al., 2015), increased NADPH/NADP
+ ratio (Clement et al., 2018), decreased activity of mitochondrial
enzymes, as well as increased oxidative stress, all are observed in older
individuals. Data on ATP changes with aging suggests that there is an
average decline of 8% per decade in ATP producing capacity in skeletal
muscles (Payne and Chinnery, 2015).
Mitochondrial DNA mutations and reduced mitophagy lead to an
increased share of dysfunctional mitochondrial subclones (Bua et al.,
2006;Payne and Chinnery, 2015;Williams et al., 2013;Yu-Wai-Man
et al., 2010;Zheng et al., 2012), which is involved in a number of
pathological processes, including macular degeneration
(Karunadharma et al., 2010) and muscle aging (Brierley et al., 1998).
Mitochondrial biogenesis is progressively impaired in aging. This
includes decline in the number of mitochondria per cell, pathologic
morphology and physiology, as well as functional changes (Bianchi
et al., 2013;Peterson et al., 2012;Vega et al., 2015). Decrease in mi-
tochondrial biogenesis in aging is aected by various types of damage
and mitochondrial defects. Peroxisome proliferator-activated receptor
gamma coactivator 1α(PGC-1α) is the master regulator of mitochon-
drial biogenesis, and age-related decline in its activators, including
AMP-activated protein kinase (AMPK), NAD-dependent deacetylase
sirtuin-1 (SIRT1), NO and cGMP, or PGC-1αdownstream eectors, such
as peroxisome proliferator-activated receptors (PPARs), estrogen-re-
lated receptors (ERRs), Nuclear Respiratory Factor 1 (NRF1) and Nu-
clear Respiratory Factor 2 (NRF2), can improve mitochondrial output
(Suliman and Piantadosi, 2016). Induction of cellular tumor antigen
p53 by cellular stress, including telomere attrition, compromises mi-
tochondrial biogenesis aecting negatively PGC-1α(Sui et al., 2016).
Mitochondrial dysfunctions and subsequent cellular apoptosis can
be caused by cell damage, calcium ions (Ca
2+
)(Giorgi et al., 2015),
ROS signalling, IR and obesity (Ptitsyn et al., 2006), excess free fatty
acids (FFAs) (Di Paola and Lorusso, 2006), or misfolded proteins (Baud
et al., 2013;Payne and Chinnery, 2015).
NAD+ deciency is mediated at least partially by an increase in
CD38 protein expression, which can be induced by inammation, toxins
(Chini et al., 2018), ROS, and endogenous vasoconstrictors (Lee et al.,
2015). Overactivation of poly (ADP-ribose) polymerase (PARP) due to
DNA damage and repair can also reduce NAD+ levels. Age-related
reductions in nicotinamide phosphoribosyltransferase (NAMPT) levels
is another possible source of NAD+ deciency (Chini et al., 2018).
ATP shortage is associated with muscle (Marzetti et al., 2013) and
cardiac (Yaniv et al., 2013) aging, and possibly with proteostasis fail-
ures (Brehme et al., 2014).
Aging-related defects of mitochondrial respiration include
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
6
pathological changes in the turnover of ROS, that are naturally occur-
ring byproducts of mitochondrial metabolism. They also play a role in
immune defense and act as signaling molecules. At the same time,
oxidative stress plays signicant roles in aging, many of which have yet
to be investigated. However, some pro-longevity interventions, such as
physical exercise (Radak et al., 2013) or caloric restriction (Nisoli et al.,
2005) lead to increased mitochondrial biogenesis and, as a result,
higher ROS levels, which is, however, combined with improved anti-
oxidant defense. Therefore, dysregulated ROS metabolism, including
the impairment of antioxidant defense and oxidation product clearance
systems lead to the increased ROS burden (Guest et al., 2014;Massudi
et al., 2012) and health deterioration seen in aging (Payne and
Chinnery, 2015).
ROS are implicated in the development of cardiovascular (Gracia
et al., 2017;Puca et al., 2013) and neurodegenerative diseases (Kim
et al., 2015a), aging of cartilage tissues (Ye et al., 2017), liver (Zhang
et al., 2014a) and bladder (Camões et al., 2015), brosis, cancer (Morry
et al., 2017) as well as fertility decline in men (Gunes et al., 2016) and
women (Briley et al., 2016).
The burden of ROS, inammation, cellular senescence (Calhoun
et al., 2016), immune senescence, oxidative stress (Lowery et al., 2013),
and misfolded protein damage (Baud et al., 2013) all promote the in-
duction of mitochondrial transition pore permeability. Mitochondrial
membrane impairment causes inadequate Ca
2+
signalling, which leads
to cell apoptosis and contributes to the development of cardiac (Dai
et al., 2012), neurodegenerative (Liao et al., 2017), muscular, cardiac
(Dai et al., 2012), and eye (Gauthier and Liu, 2017) pathologies.
Estrogen aects mitochondria breathing chain through the estrogen
receptors and NRF1, and is an important cofactor of mitochondrial
antioxidant defense, and it is thought to be the main reason of relatively
higher lifespan in females (Viña et al., 2005). Estrogen levelsdecrease
in women after menopause has signicant deteriorating health eects
(Jones and Boelaert, 2015).
A number of mitochondrially addressed interventions may amelio-
rate aging-relation conditions.
Exercise (Wang et al., 2011), sildenal(Li et al., 2018), quercetin
(Nieman et al., 2010) and epicatechin (Ramirez-Sanchez et al., 2013)
supplementation may improve mitochondrial biogenesis.
Nicotinamide riboside supplementation improves NAD + blood
levels (Airhart et al., 2017;Trammell et al., 2016;Yoshino et al., 2018),
while its clinical relevance is being investigated.
There is some promising evidence for a direct eect of the NAD+
precursor acipimox on muscle mitochondrial function in humans in-
cluding increased insulin sensitivity, and increased mitochondrial re-
spiration in skeletal muscles (van de Weijer et al., 2015). Additionally,
acipimox can reduce plasma glucose, improve blood lipid prole, pre-
vent cardiac parasympathetic modulation in hypopituitary men
(Vestergaard, 2017).
A number of antioxidant interventions are eective in reducing
mortality in some groups of patients, including glucosamine and
chondroitin (Katoh et al., 2017), CoQ10 (Alehagen et al., 2018), pen-
toxifylline (McCarty et al., 2016), NAC (Darweesh et al., 2017), mela-
tonin (Raygan et al., 2017), edaravone (Okada et al., 2018), SGLT2
inhibitors (Fatima et al., 2017;Shigiyama et al., 2017), anti-RAAS drugs
(Baykal et al., 2003), curcumin (Tabrizi et al., 2018b), resveratrol
(Tabrizi et al., 2018a) and olive oil phenols (Fabiani, 2016). Melatonin
reduces cancer mortality (Seely et al., 2012).
Other promising anti-oxidant interventions include MitoQ (Rossman
et al., 2018), carnosine (Regazzoni et al., 2016), and the intake of
polyphenols like avonoids, such as quercetin (Askari et al., 2012),
procyanidins (Li et al., 2015), and stilbenes (Reinisalo et al., 2015).
Coee consumption (Martini et al., 2016), vitamin D (Farrokhian et al.,
2017), S-adenosylmethionine (SAMe) (Loguercio et al., 1994), gluco-
samine (Katoh et al., 2017) also have antioxidant eects. Statin use has
ambivalent eects on mitochondrial metabolism (Bouitbir et al., 2012).
Interestingly, mitochondrial transplantation has not only shown
promising results in animal studies, but also has been used in humans to
improve assisted reproduction outcomes (Woods and Tilly, 2015).
3.2.7. Insulin resistance
Regulation of cellular glucose and other nutrient metabolism is
necessary for organismal functionality (López-Otín et al., 2013).
Insulin resistance (IR) plays a major role in critical health condi-
tions, such as inammation and starvation. However, impaired insulin
sensitivity is a major hallmark and an early sign of aging (Belsky et al.,
2015).
IR is a pathological condition in which cells fail to respond to insulin
normally. Signicant number of middle-aged and older adults have
diabetes or prediabetes (Yip et al., 2017). IR is associated with higher
risks of cardiovascular, kidney diseases and all-cause mortality (Chan
et al., 2017;Huang et al., 2016b). Diabetes is associated with increased
risks of diabetic nephropathy, diabetic retinopathy, cognitive decline
(Cheng et al., 2012), dementia, mobility decline, falls (Lu et al., 2009),
and some cancers (Xu et al., 2017;Zhang et al., 2017). It is char-
acterized by impaired glucose utilization, high blood sugar, increased
lipolytic activity in fat cells, circulation of FFAs and inappropriate ac-
cumulation of lipids, including abdominal and liver fat.
Chronic β-cells exposure to FFAs may lead to the loss of insulin
response by β-cells, which eventually may become permanent
(Mookerjee et al., 2010).
A number of factors are associated with IR, including inammation
(Park et al., 2014), pro-brotic signaling (Lark and Wasserman, 2017),
oxidative stress (Drougard et al., 2015), exposure to ectopic lipids such
as FFAs and to some misfolded proteins (Blaise et al., 2013;Singh et al.,
2015;Taylor et al., 2015). Excess body weight, lipid accumulation in
insulin-targeted organs including skeletal muscles and liver, and hor-
monal aging are all associated with IR, which can be exacerbated by
suboptimal diet, inactivity and a sedentary lifestyle (Shulman, 2014),
but also with inammation (Park et al., 2014) and the history of mal-
nutrition (Francis-Emmanuel et al., 2014). Dysfunctions of hypotha-
lamic tissues (Drougard et al., 2015), as well as dysregulated adipokine
signaling (Jaganathan et al., 2018) play major roles in IR development.
In fact, IR promotes LDL resistances (via proprotein convertase
subtilisin/kexin type 9 (PCSK9)-centered pathways which involves
sterol regulatory element-binding transcription factor 1 (SREBP1),
glucokinase, and hepatocyte nuclear factor 1 (HNF1)) (Miao et al.,
2015;Wiciński et al., 2017;Zhang et al., 2016). Similarly, impaired
fatty acid beta-oxidation is associated with IR.
Fatty acid resistance (inadequate fatty acid beta-oxidation) can be
triggered by overfeeding, aging-related mitochondrial, adipocyte and/
or hepatocyte dysfunction, as well as by IR (Kim et al., 2006). This
results in the elevated plasma FFA levels commonly seen in aging
(Pararasa et al., 2015). Increased systemic FFA levels (saturated FFAs in
particular) increases the risks of allcause and cardiovascular mortality
and diseases, such as NASH and NADFL, as well as high blood pressure,
impaired endothelial cell function, increased inammatory markers,
mitochondrial aging (Gaddipati et al., 2010;Pararasa et al., 2015) and
renal brosis (Kang et al., 2014). Fatty acid resistance involves (in
addition to IR) altered signaling of adipokines (Jura and Kozak, 2016),
peroxisome proliferator-activated receptor δ(PPARδ), pyruvate dehy-
drogenase lipoamide kinase isozyme 4 (PDK4) (Kim et al., 2006), solute
carrier family 13 member 5 (mINDY) (Rogina, 2017), FAT3, CD36, and
long-chain fatty acyl-CoA (LCFA-CoA) (Pararasa et al., 2015).
Elevated plasma triglycerides can result from overeating, IR, general
lipid abundance in the organism and high FFA levels (Welty, 2013).
Hypertriglyceridemia is involved in the development of NASH (Amir
and Czaja, 2011) and pancreatitis, and is associated with increased risks
of cardiovascular and all-cause mortality (Liu et al., 2013).
Resistance to the anabolic action of insulin and increased adiposity
are directly and mechanically involved in anabolic resistance, i.e. aging-
associated decrease in protein synthesis in the presence of adequate
amount of protein in food, which results in muscle loss and sarcopenia
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
7
in older people. Major factors of anabolic resistance include aging, in-
activity, obesity (Smeuninx et al., 2017), IR (Cleasby et al., 2016), and
inammation (Xia et al., 2017).
Therefore, progressive resistance to many essential nutrients such as
glucose, lipids and proteins is observed in aging, which is more pro-
minent in inactive or obese older individuals. IR plays a major role in
this impaired nutrient sensing.
A number of interventions can improve IR providing major health
benets, including exercise (Conn et al., 2014), avoiding prolonged
sitting (Benatti and Ried-Larsen, 2015), weight loss (Houmard et al.,
2002) and weight-loss inducing interventions (Khera et al., 2018;
Silvestre et al., 2018), healthy diets (Schwingshackl et al., 2018a,
2017a), consumption of olive oil (Schwingshackl et al., 2017b), whole
grains (Marventano et al., 2017), nuts (Kim et al., 2018c) and coee as
well as supplementation with polyphenols, including avonols
(Menezes et al., 2017) and anthocyanins (Yang et al., 2017).
Some antidiabetic medications signicantly reduce all-cause and
cardiovascular mortality in people with diabetes, including metformin
(Haukka et al., 2017), glucagon-like peptide-1 (GLP-1) agonists, SGLT2
inhibitors (Fatima et al., 2017). In fact, metformin use is also associated
with cancer-specic mortality reduction (Haukka et al., 2017).
A number of anti-glycemic interventions decreases not only plasma
glucose but also triglyceride or FFA levels (Rosenblit, 2016), for ex-
ample, metformin reduces the levels of plasma FFAs (Castro Cabezas
et al., 2012).
Other anti-glycemic and anti-triglyceridemic interventions include
acipimox (Makimura et al., 2016), ginger (Mazidi et al., 2016), pro-
biotics (Kasińska and Drzewoski, 2015;Wu et al., 2017), berberin (Lan
et al., 2015a), vitamin D (Jafari et al., 2016;Poolsup et al., 2016), olive
leaf extract (de Bock et al., 2013;Lockyer et al., 2017), magnesium
(Verma and Garg, 2017), sildenal(Ramirez et al., 2015), melatonin
(Doosti-Irani et al., 2018), fenugreek (Gong et al., 2016), Salacia
(Jeykodi et al., 2016), Tribulus Terrestris (Samani et al., 2016), As-
tragalus (Tian et al., 2016) and carnosine (de Courten et al., 2016;
Menon et al., 2018).
3.2.8. Aging-related body composition changes
Aging in humans is associated with a decreased share of lean mass
and an increased share of fat body mass, adipose tissue expansion, and
ectopic fat deposits in visceral area, muscles, heart, blood vessels, bone
marrow, liver, and pancreas.
The decline in non-fat cell mass in aging goes together with de-
creased mass of muscles, spleen, liver, kidney, brain, and bones
(Manini, 2010). Lung volume and functionality is decreased in older
ages (Koch et al., 2011).
Age-related decline in maximal oxygen uptake (VO2max) results
from multiple factors: decreased maximal heart rate and stroke volume,
stiening heart muscle bers, reduced muscle volume and strength, and
as well as diminished blood volume (Kim et al., 2016a).
Skeletal muscle mass usually contributes up to about 50% of total
body weight in young adults in high-income societies and decreases
during aging to approximately 25% of total body weight by 7580 years
(Short et al., 2004), Skeletal muscle loss has major negative impact on
indicators of whole body mitochondrial capacity, VO2max (Coen et al.,
2013;Mathers et al., 2012) and resting metabolic rate (RMR). Skeletal
muscle loss was detected in both aged untrained individuals and pro-
fessional athletes (Kusy and Zieliński, 2014), but it is greatly ex-
acerbated by low physical activity.
Modern humans evolved from hunting and gathering societies,
where people were used to very high physical activity levels throughout
old age. Traditional farmers also have rather high physical activity
compared to modern oce workers. Food has never been as abundant
as in the last few decades in the developed countries, and human brains
are not tted for rigorous appetite control, as our ancestors had to
survive through persistent risks of malnutrition and frequent famines.
As a result, humankind faces new challenges with detrimental health
eects of sedentary lifestyle and obesity (Kirchengast, 2014).
Additionally, aging-related muscle loss is mediated by IR, anabolic
resistance, epigenetic reprogramming, and mitochondrial dysfunctions
including Complex I dysfunction, reduced ATP synthesis (Su et al.,
2015), reduced muscle oxygen consumption (Coen et al., 2013), in-
creased ROS signalling (Doria et al., 2012), diminished Na/K-ATPase,
as well as anabolic hormone signalling, muscle denervation (Spendi
et al., 2016), and muscle brosis due to TGF-β1and myostatin signal-
ling (Parker et al., 2017).
Reduced volume of skeletal muscles and diminished lung function
aect each other negatively during aging (Koch et al., 2011). Numerous
population studies have documented an inverse association between
lung volume markers (VO2max, forced expiratory volume in 1 second
(FEV1)) and aging-related endpoints including all-cause and cardio-
vascular mortality, cognitive functions, T2D (Zaccardi et al., 2015), and
fractures (Kim et al., 2018c;Mathers et al., 2012).
In addition to decreased oxygen demand, lung volume can be ne-
gatively aected by thoracic cage stiening from rib cage calcication,
age-related kyphosis from osteoporosis, persistent low-grade in-
ammation, proteolytic and oxidant-mediated injury to the lung matrix
resulting in the loss of alveolar units, elastin loss, as well as with im-
mune senescence and loss of lung stem cell regenerative capacity
(Brandenberger and Mühlfeld, 2017).
Aging-related loss of lean body mass can result in either body mass
loss or increased share of adipose tissue. Both obesity and being un-
derweight are associated with increased risks of all-cause mortality,
with low body weight being particularly dangerous in older adults
(Global BMI Mortality Collaboration et al., 2016).
Diminishing energy expenditures, RMR and physical activity levels
lead to lower energy demand. On average, people decrease their caloric
consumption during aging, which increases the risks of nutritional de-
ciencies in old age (Giezenaar et al., 2016). Many aging individuals,
however, do not adjust their caloric consumption to decreased energy
demands which results in weight gain.
Age-associated weight gain is a well documented phenomenon ty-
pically associated with aging through middle ages (Belsky et al., 2015).
Obesity itself has been shown to lead to the various forms of cellular
and tissue dysfunctions characteristic of aging, and is associated with
an increased risk of comorbidities, including diabetes, CVDs, stroke,
cancer, liver diseases, osteoporosis, osteoarthritis, and depression
(Abdelaal et al., 2017).
At the same time, aging-related increase in adiposity is causally
implicated in age-related inammation, due to inammatory marker
overexpression in obese individuals (Hotamisligil et al., 1997;Weisberg
et al., 2003). Similarly, aging-related weight gain and obesity are the
major factors of IR, leading to further weight gain and insulin in-
sensitivity (Pararasa et al., 2015).
A large share of the middle-aged and older overweight individuals
persistently try, but fail to control their appetite and food consumption,
which might be aggravated by aging-related conditions such as IR,
adipokine signalling changes, as well as by reduced signalling of neu-
rotransmitters, including dopamine and gamma-Aminobutyric acid
(GABA) (Delgado, 2013). Uncompensated overeating is the major
source of aging-related weight gain and obesity.
Eating behavior and physical activity are inuenced by various
neurotransmitters and changes in their metabolism in aging. Aging-
related decline in physical activity is detrimental to both muscle ca-
pacity and insulin tolerance. It is observed not only in older humans,
but also in other species. It is determined by aging-related changes in
neurotransmitter signalling, including orexin, dopamine, and agouti-
related protein (AGRP) (Knab and Lightfoot, 2010;Manini, 2010;
Ruegsegger and Booth, 2017).
Similarly, hormones and other neurotransmitters have major eects
on appetite and the ability to control food intake, including insulin
sensitivity mediator signalling, such as adipokines (Koleva et al., 2013),
dopamine, GABA (Delgado, 2013), neuropeptide Y, AGRP (Ferrario
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
8
et al., 2016), brain-derived neurotrophic factor (BDNF), ciliary neuro-
trophic factor (CNTF), α-melanocyte-stimulating hormone (α-MSH),
and a lot of these signals are diminished or altered in aging (Xu and Xie,
2016). Hypothalamic gliosis and TGF-βsignalling is associated with IR
and obesity, which implies the role of neurodegeneration in these
processes (Dorfman and Thaler, 2015;Schur et al., 2015).
Increased adipocyte volume in obese individuals is associated with
the inability to store triglycerides, impaired mitochondrial function,
alterations in membrane proteins and higher levels of adipocyte cell
death and inammation, all of which have causal implications in the
development of obesity-related metabolic dysfunction (Heinonen et al.,
2014;Maurizi et al., 2018). Larger hypoxic areas of the adipose tissues
in combination with its increased proinammatory microenvironment,
as seen in age-related increase in adiposity, is further aggravated by IR
(Cinti et al., 2005;Kovsan et al., 2011).
Aging promotes the shift from subcutaneous and lower body adip-
osity to visceral fat expansion, which has detrimental metabolic eects,
including inammation, IR, cardiovascular diseases (Manini, 2010;
Pararasa et al., 2015) and NAFLD (Verrijken et al., 2010). Visceral
adipose tissue secretes more pro-inammatory cytokines, promotes IR
with lower adiponectin signalling and leads to higher FFA plasma levels
due to its lower fat storing capacity compared to subcutaneous fat depot
(Manini, 2010;Pararasa et al., 2015). Enlarged visceral adipocytes are
associated with dyslipidemia (Hostedt et al., 2010).
Liver fat deposits are the main cause of NAFLD and NASH, con-
tributing to liver brosis and cirrhosis. Liver volume decreases by 20-
40% as a person gets old, predominantly due to functional cell loss (Kim
et al., 2015b). Major mechanisms linked to liver fat accumulation in-
clude cellular senescence, which leads to impaired liver regeneration,
obesity, inammation, hypoxia, IR, and brosis (Pararasa et al., 2015).
Mitochondrial dysfunctions also promote the hepatic fat expansion,
including SIRT1 and NAMPT (Gaddipati et al., 2010) loss and oxidative
stress (Masarone et al., 2018). Toxicant exposure and brosis can lead
to reduced number and size of fenestrations (pores) of liver sinusoidal
endothelial cells. Liver defenestration may lead to an increase in liver
fat deposits and impaired lipid processing in the liver (Kim et al.,
2015b).
Overeating leads to increased plasma lipid levels, and together with
oxidative stress and inammation promotes atherosclerosis develop-
ment.
Increased share of fat in bone marrow aects bone health negatively
leading to lower bone density with increased risks of osteoporosis and
vertebral fractures (Schwartz, 2015).
Physical activity, including aerobic and resistance exercise, is at
least partly capable of preventing and restoring aging-related loss of
muscle mass, energy expenditure (Siparsky et al., 2014), and VO2max
(Burtscher, 2013).
A number of nutritional supplements have been used to protect
muscle mass and function and promote physical activity and mobility in
older people, including vitamin D3 (Cleasby et al., 2016), higher caloric
content, amino acids, high protein content (Cawood et al., 2012), n-3
fatty acids (Bell et al., 2017), and β-Hydroxy β-Methylbutyrate (HMB),
a metabolite of leucine amino acid (Wu et al., 2015b). A high-protein
oral nutritional supplement with HMB reduced mortality among mal-
nourished, older, hospitalized adults (Deutz et al., 2016).
Testosterone supplementation for men alone or in combination with
human growth hormone (GH) might increase muscle mass and decrease
fat mass (Giannoulis et al., 2012). However, the safety of these ap-
proaches is still a major issue, as pro-anabolic signalling is implicated in
the development of aging-related diseases (for example, insulin-like
growth factor 1 (IGF-1) and mTOR signalling activated by GH, as well
as dihydrotestosterone, a testosterone metabolite, are involved in both
normal regenerative processes in neural or muscle tissues, but also in
aging and disease development (Saxton and Sabatini, 2017). Another
example is dehydroepiandrosterone (DHEA) supplementation, which
improves body composition in older men (Corona et al., 2013), but
whose safety is still not clear.
Certain diets can promote weight reduction in overweight in-
dividuals, including energy restriction, low-fat, low-carbohydrate,
Mediterranean (Mancini et al., 2016), and vegetarian (Huang et al.,
2016a) diets, nut and legume consumption (Schwingshackl et al.,
2018b). Exercise can also be eective for weight loss if combined with a
proper diet (Stoner et al., 2016).
A number of pharmacological interventions are eective at pre-
venting, slowing and reversing both weight-gain in general as well as
age-related weight gain. Some antidiabetic drugs (in addition to redu-
cing morbidity and mortality) are found to reduce body weight in
people with diabetes: metformin, acarbose, miglitol, pramlintide, ex-
enatide, liraglutide (Domecq et al., 2015). The use of liraglutide as an
anti-obesity agent is currently studied for long-term health eects
(Davies et al., 2018).
Resveratrol (Mousavi et al., 2018b), probiotics (Zhang et al., 2015),
curcumin (Mousavi et al., 2018a) and ginger (Maharlouei et al., 2018)
are somewhat eective for reducing IR and for weight loss, but re-
sveratrol use might increase LDL (Zhao et al., 2019a).
Bariatric surgeries provide major weight loss benets and lead to
lower long-term all-cause cardiovascular and cancer mortality among
young and middle-aged adults with obesity (Cardoso et al., 2017).
Orlistat reduces fat absorption, and consequently, caloric con-
sumption. This results not only in weight loss, but also in improved
blood lipid proles (Sahebkar et al., 2017).
A number of medicines lead to weight reduction as direct or side
eects, aecting neurotransmitters, such as dopamine, GABA, gluta-
mate, and serotonin: phentermine, zonisamide, topiramate, bupropion,
uoxetine, phentermine, lorcaserin (Domecq et al., 2015). Long-term
safety of these agents remains to be explored. Phentermine/topiramate
controlled-release use was associated with weight loss, improved pa-
tients' lipid proles, particularly their levels of triglycerides and HDL
cholesterol, insulin sensitivity and glycemia, retarded the progression of
T2D (Bays and Gadde, 2011;Kiortsis, 2013), but tended to increase
heart rates (Alfaris et al., 2015).
Statins signicantly reduce not only cardiovascular risks but also
all-cause mortality in people with dyslipidemia (Naci et al., 2013). Yet,
statin use might cause myopathy as a side eect.
Lipid-lowering interventions with potential health benets are
berberine (Lan et al., 2015b), axseed interventions (Pan et al., 2009),
consumption of ginger (Mazidi et al., 2016), olive products (Filip et al.,
2015), dietary avonoid intake (Menezes et al., 2017), and policosanol
(Gong et al., 2018).
3.2.9. Aging-related hormonal changes
Hormone proles change signicantly in aging humans, and many
of these changes are clinically signicant, leading to health impairment
and frailty.
Aging brings reduced production of sex hormones, which leads to
physiologic changes: reduced muscle mass, energy, and exercise capa-
city, bone loss, increased risks of AD (Jones and Boelaert, 2015;Uchoa
et al., 2016) and decline in sexual function (Williams and Cho, 2017).
Aging-related estrogen loss is especially pronounced in women after
menopause, which has direct detrimental health eects. Estrogen re-
ceptors are important transcriptional factors regulating pathways of
mitochondrial oxidative metabolism, which might partly explain lower
mortality in females compared to males (Austad and Fischer, 2016).
A later age of natural menopause (ANM) correlates with lower all-
cause mortality (2% with each increasing year of ANM), decreased risk
of CVD, atherosclerosis, stroke and osteoporosis (Schoenaker et al.,
2014). These benets seem to outweigh side eects such as a moderate
increase in breast and ovarian cancer risks (Day et al., 2015;
Schoenaker et al., 2014). Menopause-related estrogen loss increases
cardiovascular risk factors, such as endothelial dysfunction, dyslipi-
demia, hypertension, IR, hypercoagulability, and pro-inammatory
state. It also increases risks of osteoporosis (Jones and Boelaert, 2015),
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
9
and leads to changes in body composition, neurodegeneration (Uchoa
et al., 2016) and optic nerve aging (Dewundara et al., 2016).
Low levels of endogenous testosterone in older men are associated
with increased risks of all-cause and cardiovascular mortality (Araujo
et al., 2011) and AD (Lv et al., 2016), as well as higher fat mass (Bann
et al., 2015), metabolic syndrome, diabetes, dyslipidemia, hyperten-
sion, renal failure, frailty, malignancy and cardiovascular events
(Shores and Matsumoto, 2014).
The blood levels of DHEA and its metabolite dehydroepian-
drosterone sulfate (DHEAs) decrease in aging. Low DHEA and DHEAs
levels in older people are associated with increased risks of mortality
(Rosero-Bixby and Dow, 2012), frailty (Tajar et al., 2011), depression,
T2D and AD (Jones and Boelaert, 2015).
In summary, reduced sex hormone metabolism is associated with
generally poorer health in aging individuals.
Aging inuences many aspects of complex hormonal system. The
cells and tissues of the neuroendocrine glands, including hypothalamus,
a brain structure that controls the other structures in the endocrine
system, are aected by aging-related neurodegenerative processes.
In turn, neural aging is driven by pathological processes like neu-
roinammation, microglia excessive activation, cellular senescence
(Ojo et al., 2015), mitochondrial dysfunction, oxidative stress, ac-
celerated neuronal apoptosis, impaired neurogenesis, neurovascular
aging, gliosis, and IR (Hardeland et al., 2015;Yin et al., 2016). Im-
paired proteostasis, including impaired amyloid processing, exacer-
bates neural aging, including the aging of neuroendocrine organs re-
ducing brain neurosteroid synthesis among other things (Barron and
Pike, 2012).
There is preclinical data that inammatory signalling, such as TNF-
α, inhibitor of nuclear factor kappa-B kinase subunit beta (IKK-β) and
NF-κB, impairs hypothalamic neurogenesis leading to aging-related
decline in gonadotropin-releasing hormone (GnRH) signalling (Zhang
et al., 2013). Hypothalamic gliosis is associated with IR and obesity
(Schur et al., 2015), which could be signs of inammatory response
(Dorfman and Thaler, 2015;Schur et al., 2015). Hypothalamic aging
results in decrease and dysregulation of GnRH pulsations (Veldhuis
et al., 2009;Zhang et al., 2013), which can be detrimental for sex
hormone metabolism. In fact, about 85% of male hypogonadism cases
are secondary in the sense that the testes are being insuciently sti-
mulated by the hypothalamic-pituitary axis (McCullough, 2015).
Also, neurodegeneration of hypothalamic suprachiasmatic nucleus
is involved in reduced melatonin metabolism, which causes sleep dis-
turbances and deprives aging human organisms of multiple benets of
melatonin, including anti-inammatory and antioxidant defense and
bone health support (Hardeland et al., 2015). In addition, the size of
melatonin-producing pineal gland decreases in aging.
At the same time, human gonads (testes and ovaries) are aected by
aging as well. Aging testicular tissues undergo volume loss, IR, oxida-
tive stress, vascular dysfunctions, hypoxia and brosis. Testosterone-
producing Leydig cells suer from mitochondrial dysfunction and oxi-
dative stress resulting in reduced steroidogenesis (Wang et al., 2017).
They decrease in quantity, which plays a role in aging-related reduction
in androgen levels (Gunes et al., 2016).
Menopause involves follicular dysfunctions and depletion, as well as
hypothalamic function alterations. The number of primordial follicles
rapidly declines with time. About 1 million oocytes are present at birth
in the human ovary, about 400,000 follicles are present at puberty, and
only about 500 (about 0.05%) ovulate, while the rest are wasted
(Wilkosz et al., 2014).
Decreased number and function of follicular granulosa cells results
in decreased estrogen and inhibin B secretion. This leads to increased
GnRH and follicle-stimulating hormone (FSH) secretion during early
stage of menopause (Harlow et al., 2012). Additionally, the signaling of
hypothalamic GnRH-promoting peptide kisspeptin is signicantly in-
creased with womens age which accelerates menopause (Day et al.,
2015). FSH promotes growth and recruitment of immature ovarian
follicles, which ultimately accelerates their depletion, leading to me-
nopause (Day et al., 2015).
Primordial and small follicle loss with aging results in the loss of
inhibin B and Anti-Müllerian hormone, as they are secreted by smaller,
less developed follicles (Rojas et al., 2015). This results in accelerated
oocyte development to a more mature state which makes them more
prone to apoptosis and follicular atresia. Depletion of follicles leads to
signicant decrease in androgen and estrogen production, declining
fertility and ultimately menopause. GnRH secretion is diminished
during late postmenopause (Harlow et al., 2012).
Oocytes suer from reduced oxidative defense, mitochondrial dys-
functions, telomere attrition, lipotoxicity (Diamanti-Kandarakis et al.,
2017;Keefe et al., 2015), AGEs, and DNA damage (Wilding, 2015).
Ovarian DNA damage repair system deteriorates with age, as its prin-
cipal agent breast cancer type 1 susceptibility protein (BRCA1) is pro-
gressively lost in aging (Oktay et al., 2015) which is related to meno-
pause (Day et al., 2015). Additionally, ovarian stromal cells and
adjusting tissues undergo cellular senescence which leads to increased
inammatory signalling, tumorigenic propensity (Shan and Liu, 2009),
and ovarian stromal brosis (Briley et al., 2016).
Testosterone replacement therapy improves health, quality of life
and reduces all-cause mortality in men with hypogonadism (Hackett,
2016). It has benets in terms of sexual and physical functioning, mood,
and depressive symptoms, however, its long-term safety needs to be
evaluated in large clinical trials (Snyder et al., 2016). At the same time,
recreational anabolic steroid use by young men leads to prolonged
decrease of endogenous serum testosterone levels (Christou et al.,
2017). There is early clinical data on hypogonadism treatments, cap-
able of restoring endogenous testosterone production in men, including
human chorionic gonadotropin, clomiphene citrate, and some ar-
omatase inhibitors (McCullough, 2015).
Certain preparations of HRT may provide signicant symptom-re-
ducing benets for younger healthy postmenopausal women (5060
years old). The benets include a reduction in all-cause mortality,
coronary health disease and osteoporotic fractures. At the same time,
certain HRT regimes can increase risks of breast, ovarian and uterine
cancers, and thrombotic events, which limits HRT use (Lobo, 2017).
Further clinical studies are needed.
Nulliparity, vegetarian diet, smoking, high polyunsaturated fat in-
take, and excessive exercise consistently correlate with earlier age at
natural menopause. Higher BMI, higher intake of total fat, protein, and
meat, dietary vitamin D and calcium intake, as well as moderate phy-
sical activity and exercise delay the age at natural menopause (Sapre
and Thakur, 2014).
Coenzyme Q10 supplementation might preserve female fertility, but
further research is needed (Gretchen Garbe Collins, 2015).
3.2.10. Cardiovascular aging
Cardiovascular diseases (CVDs) are a major cause of aging-related
morbidity and mortality. Ideal cardiovascular health metrics are asso-
ciated with a 46% lower all-cause mortality compared to people with
the worst indicators (Guo and Zhang, 2017).
By delivering oxygenated blood to all tissues in the body, the car-
diovascular system plays a vital role in the health and longevity of the
organism as a whole. Accumulation of cardiovascular system dysfunc-
tions can make cardiovascular system a major driver of pathological
processes in an aging individual. Therefore, we denote cardiovascular
aging as a separate and major causal process in aging pathogenesis.
Thus, peripheral artery disease can lead to stroke, renovascular
hypertension, renal dysfunctions, mesenteric ischemia, ischemia in
lower and upper extremities (Aboyans et al., 2018). Cerebrovascular
dysfunctions are major risk factors for stroke, dementia, cognitive de-
cline, and neural aging (Toth et al., 2017). Vascular aging and diseases
are involved in the pathogenesis of eye diseases, including macular
degeneration, Schnabel cavernous degeneration, and possibly, presby-
opia (Pescosolido et al., 2016), as well as loss of hearing (Kurata et al.,
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
10
2016) and taste sensitivity (which could lead to malnutrition in old age)
(Pavlidis et al., 2013). Microvascular damage is involved in many pa-
thological processes, including neural, myocardial, lung, kidney, colon,
skin (Ambrose, 2017), and tendon aging (McCarthy and Hannan,
2014). Also, vascular damage is a major cause of erectile dysfunction
(El-Sakka, 2011).
Aging has a remarkable eect on the heart and vascular system,
leading to the increase in CVD incidence including ischemic heart dis-
ease, atherosclerosis, hypertension, heart failure, myocardial infarction,
and stroke. Cardiovascular tness (measured, for example, by VO2max)
declines in aging adults, especially in people with low physical activity
(Burtscher, 2013).
Cardiac and vascular, in particular, arterial wall remodeling, stif-
fening and brosis seem to be major processes contributing to aging
pathogenesis as a whole. Vascular remodelling sincludes irreversible
degradation of elastic ber in the blood vessel walls, collagen deposi-
tion and cross-linking, blood vessel wall thickening, amyloid deposition
in the medial layer, and migration/proliferation of vascular smooth
muscle cells to the intima. Heart aging can be exacerbated by left
ventricular (LV) hypertrophy, altered LV diastolic function, diminished
LV systolic reserve capacity, increased arterial stiness, impaired en-
dothelial function and cardiac rhythm disturbances (North and Sinclair,
2012). Cardiovascular aging is exacerbated by hypertension, dyslipi-
demias, ischemia, reduced vascular perfusion (Ungvari et al., 2018),
and altered noradrenergic vascular innervation (Hunter et al., 2012).
It is clear that low physical activity greatly contributes to en-
dothelial dysfunction and aggravates cardiovascular aging (de Almeida
et al., 2017). There is evidence of people from a few modern hunter-
gatherer and forager-horticulturalist societies having unusually low
prevalence of atherosclerosis implying a major role of lifestyle in car-
diovascular aging (Kaplan et al., 2017;Lemogoum et al., 2012).
Both menopause and hypogonadism have particular detrimental
eects on cardiovascular health (Jones and Boelaert, 2015).
The pathological processes of cardiovascular aging are driven by
mechanical stress in the blood vessel walls, exacerbated by hyperten-
sion and RAAS activation, pro-inammatory, pro-brotic (TGF-β) and
proteolytic (MMP-12) signalling, cellular senescence, mitochondrial
dysfunction, IR, immune senescence (Yu et al., 2017), as well as by
misfolded proteins such as amyloids, AGEs, AOPPs, and elastin peptides
(Hunter et al., 2012;Ungvari et al., 2018).
Pathogenesis of atherosclerosis involves dysregulated cholesterol
metabolism, oxidation of TG-rich lipoproteins, cellular senescence, in-
ammation (IL-1, TNF-α, IL-6 pathways) (Ridker and Lüscher, 2014),
and mitochondrial dysfunctions (Ungvari et al., 2018). Dysfunctional
calcium signalling may lead to both excessive cellular apoptosis and
blood vessel calcication. Vitamin D deciency is associated with
higher all-cause, cardiovascular and cancer mortality, respectively
(Chowdhury et al., 2014).
Dysregulation of nitric oxide (NO) synthesis and processing occurs
with age and in the context of CVDs due to inammation and oxidative
stress (Ungvari et al., 2018).
A number of interventions are eective for primary or secondary
prevention of cardiovascular aging, diseases, as well as of cardiovas-
cular and all-cause mortality.
Physical activity and exercise, as well as reducing daily sitting time,
have major potentials in primary and secondary prevention of CVDs
(Alves et al., 2016), and some cancers (Kruk and Czerniak, 2013).
Healthier dietary patterns are associated with decreased cardio-
vascular and all-cause mortality, including whole-grain food, fruit, ve-
getable, nut, legume, and sh consumption, certain polyphenols (Kim
et al., 2018c;Rienks et al., 2017), moderate coee intake (Zhao et al.,
2015), and possibly olive oil (Schwingshackl and Homann, 2014).
Excessive consumption of red meat, especially processed meat, and
sugar-sweetened beverages have negative health eects (Bechthold
et al., 2017;Schwingshackl et al., 2017c). Similar associations are ob-
served for cancer risks (Bella et al., 2017).
Anti-hypertensive drugs reduce not only blood pressure, but also all-
cause mortality, CVD events, stroke, and heart failure in people with
hypertension (Ettehad et al., 2016), as well as retinopathy progression
(Xie et al., 2016).
Statin use in people at increased CVD risks (Chou et al., 2016), as
well as the use of some antidiabetic medications in people with T2D
(Haukka et al., 2017), such as metformin (Campbell et al., 2017), GLP-1
agonists and SGLT2 inhibitors reduce cardiovascular and all-cause
mortality (Fatima et al., 2017). Statin use might also decrease some
(Mei et al., 2017) cancer risks. Certain preparations of hormone re-
placement therapy, including parenteral estrogen therapy for younger
postmenopausal women (Lobo, 2017), as well as, possibly, long-acting
testosterone injection supplementation for men with hypogonadism
(Hackett, 2016) reduce cardiovascular and all-cause mortality.
Some anti-inammatory treatments reduce both cardiovascular and
all-cause mortality, including aspirin (Sutclie et al., 2013). Glucosa-
mine and chondroitin supplementation, in addition to being used as
anti-osteoarthritic remedy, is associated with reduced all-cause, cardi-
ovascular, cancer and respiratory mortality (Bell et al., 2012) as well as
possibly colorectal (Ibáñez-Sanz et al., 2018;Kantor et al., 2016) and
lung cancer (Brasky et al., 2011) incidence. Glucosamine use might also
improve endothelial function (Katoh et al., 2017).
Bisphosphonate use improves calcium metabolism and may reduce
cardiovascular, all-cause and cancer mortality in patients with osteo-
porosis (Kranenburg et al., 2016;Van Acker et al., 2016), although the
eect on all-cause mortality was recently challenged (Cummings et al.,
2019). Some data suggests that low dose vitamin D3 supplementation
without calcium might reduce blood pressure (Golzarand et al., 2016),
but there is currently not enough data supporting cardioprotective ef-
fects of vitamin D supplementation to make any strong conclusions,
partly due to the lack of clinical trials in people with vitamin D de-
ciency with regular administration of various doses of vitamin D3, and
not combined with calcium. At the same time, cohort studies of Swedish
women (Yang et al., 2011), as well as of some patient groups (Ho et al.,
2014;Shapiro et al., 2014) imply that sun exposure can be protective
against cardiovascular and all-cause mortality, as well as some aging-
related diseases, despite the increased skin cancer incidence (Hoel
et al., 2016;Lindqvist and Landin-Olsson, 2017).
High dietary magnesium intake is associated with lower cardio-
vascular and all-cause mortality (Fang et al., 2016). Magnesium aects
homeostasis of both calcium and vitamin D. There seems to be an op-
timal ratio between calcium and magnesium intake, and suboptimal
levels of this ratio (Rosanoet al., 2016) disturb metabolism and ac-
celerate certain aging-related diseases. Magnesium supplementation
may decrease CVD risk factors (Verma and Garg, 2017).
CoQ10 supplementation decreased mortality and improves exercise
capacity in patients with heart failure (Li and Yan, 2017). NAC may
signicantly ameliorate the consequences of myocardial Infarction
(Pasupathy et al., 2017).
Interventions boosting cyclic adenosine monophosphate (cAMP), an
ATP derivative, including PDE inhibitors including PDE5 inhibitors
(Andersson et al., 2017;De Vecchis et al., 2017), pentoxifylline
(Champion et al., 2014) and coee intake (Poole et al., 2017;Zhao
et al., 2015), compensate for ATP deciencies and could reduce car-
diovascular and all-cause mortality at least in some groups of people.
3.2.11. Other mechanisms of aging
Aging is obviously not conned to the pathological processes de-
scribed above. We identied several other mechanisms of aging
throughout the course of our research literature review.
This includes DNA damage, including DNA lesions such as muta-
tions, translocations, chromosomal gains and losses, telomere short-
ening, and gene disruptions caused by the integration of viruses or
transposons (López-Otín et al., 2013). DNA damage has clinically im-
portant eects in many types of aging cells and tissues, including stem
cells (Franco et al., 2018;Jaiswal et al., 2017;Weakley et al., 2010), as
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
11
well as oocytes (White and Vijg, 2016) and spermatozoa (Gunes et al.,
2016) contributing to aging-related fertility loss. DNA damage is a
major cause of cancer and in some cases of CVD (Jaiswal et al., 2017)
development.
Aging-related epigenetic changes are also a major issue aecting
directly or indirectly every hallmark of aging. This includes hetero-
chromatin loss, the formation of senescence-associated hetero-
chromatin foci, changes in global DNA methylation, histone loss and
histone code modications, and alterations in biogenesis and functions
of non-coding RNA (Gangisetty et al., 2018;Pal and Tyler, 2016).
Changes of calcium metabolism in aging seem to be an important
albeit a complex (Cohen et al., 2018) issue as a number of interventions
improving calcium metabolism reduce all-cause mortality in aged
people (Fang et al., 2016;Golzarand et al., 2016;Kranenburg et al.,
2016;Van Acker et al., 2016). Aging-related pathologies include cal-
cium bone loss in osteoporosis, and vascular calcication, which are
correlated, at least in women (Zhang and Feng, 2017). On the other
hand, dysregulated calcium signalling is implicated in neurodegenera-
tive diseases of aging (Pchitskaya et al., 2018).
Finally, in addition to cardiovascular aging, aging of some other key
body organs and systems can drive the cascades of pathological pro-
cesses in organism as a whole. This includes aging-related damage to
liver, kidney, and nervous system. Similarly, stem cell exhaustion in
aging is another important issue driven by mitochondrial dysfunctions
(Zhang et al., 2018a), cellular senescence (Sui et al., 2016), DNA da-
mage (Beerman et al., 2014;Jaiswal et al., 2017), epigenetic changes,
as well as negative changes in the stem cell niches, including pro-in-
ammatory signalling and ROS (Chen et al., 2017a).
Yet, the complexity of aging does not seem to be indenite, as it can
be described in terms of a number of pathological processes, which can
be in many cases addressed therapeutically to the overall benet of the
patient.
We mapped connections between the major clinically signicant
pathogenetic processes of aging in humans identied in this article from
the current medical research literature which we described in the ear-
lier sections of the article (Fig. 1).
3.3. Course and outcome: a distinct pattern of development over time
Aging-related pathologies are of chronic nature. They may be alle-
viated but they do not vanish in their complexity. Many biomarkers
change consistently as people age, but some have non-linear relation-
ships with calendar age (Yashin et al., 2010) due to changes in nutrient
sensing described above. These changes in metabolism and pharmaco-
dynamics are clinically important factors that can be used to identify
specic stages of aging, which might include aging in young adults (25-
40 y.o.) (Belsky et al., 2015), middle-aged (40-60 y.o.), youngest old
(60-69 y.o.), oldest old (80 years and older) (Forman et al., 1992),
(Lehallier et al., 2019), as well as centenarians and supercentenarians.
Aging may ultimately result in death even in the absence of major
underlying diseases (causes of death). At least some centenarians die
without specic major diagnosed diseases as underlying causes of death
(Bernstein et al., 2004;Meslé and Vallin, 2017). About 10% of super-
centenarians survive until the last 3 months of life without major age-
related diseases (Andersen et al., 2012). For many of centenarians old
age or senility (Chen et al., 2019;Evans et al., 2014;Meslé and Vallin,
2017) was recorded as the underlying cause of death which may be
explained by the combined eects of aging-related decits leading to
general health deterioration in this group (Chen et al., 2019;Evans
et al., 2014;Meslé and Vallin, 2017). This adds to the observation that
pathological eects of aging cannot be entirely explained by ill eects
of aging-related diseases.
3.4. Treatment response: a known pattern of response to interventions
We identied and described above a number of interventions which
can reduce the symptoms of the major aging-related pathological pro-
cesses. Some of them are also eective against aging-related diseases,
Fig. 1. The major pathogenetic processes of aging.
Note: the dotted lines reect the relationships relevant for only some organs or body systems, while the solid lines mark the relationships relevant for human
organism in general.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
12
Table 1
Interventions against aging-related pathological processes
Intervention Inammation Cellular
senescence
Proteostasis
failures
Immune
senescence
Fibrotic
propensity
Mitochondrial
dysfunctions
Insulin
resistance
Body
composition
changes
Hormonal
changes
Cardiovascular
mortality
Cancer
mortality
All-cause
mortality
Physical activity + (+) + +* + + + (+)* + + +
Healthy diet + (+) + (+) +* + + + +- + + +
Olive products + (+) + + + + + +
Coee + +* (+)* + + ++
Nuts +++ + ++
Legume ++ +
Whole-grain products ++
Flavonoid consumption + + (+)* (+) + + + (+) (+)
Magnesium intake + + (+*) +*
Vitamine D3 supplementation + + + + (+) + (+) + +
Sun exposure (+) +- (+)
Probiotics ++ ++
Aspirin ++* +* + +
Coenzyme Q10 + +* + +* +* +*
Acipimox +* + + + (+)
Quercetin + + (+)* (+)* (+) +
Resveratrol +++-
Astragalus / TA65 + (+) (+)*
Berberine ++
Carnosine ++*+* + +
N-acetylcysteine + + (+)*
Ginger +++
Turmeric, curcumin ++* + +
Drugs against specic conditions
Statins +* +* +* +- +-* + +* +*
Metformin ++* +*+- +++
SGLT2 inhibitors +* + + + + +
GLP-1 agonists ++*++++
RAAS blocking drugs +* +* + + +* + +
Glucosamine / chondroitin + + (+) (+)* (+)
Bisphosphonates (+)* +* (+*)
PDE5 inhibitors + + +* (+) + +*
Pentoxifylline +++ ++*
HRT for women +* + +* +-* +*
Testosterone supplementation
for men with
hypogonadism
+* +* +*
Melatonin +++ +
Bariatric surgeries for people
with obesity
++ +
Note:+eective; - harmful; (+) possibly eective; +* eective in some groups of people; * harmful in some group of people.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
13
while some reduce all-cause mortality in large population groups
(Table 1).
Physical activity stands out as a nearly universal anti-aging cure, in
that it ameliorates most of the aging-related pathological processes
surveyed here (Alves et al., 2016;Arem et al., 2015;Duggal et al., 2018;
Kruk and Czerniak, 2013;Mundstock et al., 2015;Rahman and Adjeroh,
2019). Healthy diets (Bechthold et al., 2017;Mancini et al., 2016;Rae
et al., 2017;Schwingshackl et al., 2018a,2018b,2017a,2017c;Soltani
et al., 2017), high in avonoids (Kim et al., 2018c;Rienks et al., 2017),
olive oil (Schwingshackl et al., 2017b,2015;Schwingshackl and
Homann, 2014), coee (Liu et al., 2015;Martini et al., 2016;Poole
et al., 2017;Zhang and Zhang, 2018;Zhao et al., 2015), nuts (Kim et al.,
2018b), legumes (Schwingshackl et al., 2017c), whole grain products
(Marventano et al., 2017;Rienks et al., 2017) and magnesium (Fang
et al., 2016;Rosanoet al., 2016;Simental-Mendia et al., 2017;Verma
and Garg, 2017). Vitamin D and magnesium supplementation in people
with such mineral deciencies also seem to have broad-spectrum anti-
aging eects (Chowdhury et al., 2014;Farrokhian et al., 2017;
Mirhosseini et al., 2018).
Aspirin seems to be another anti-aging drug for the eligible patients
(Richman and Owens, 2017;Sutclie et al., 2013;Whitlock et al.,
2015).
A number of drugs need to be tested as promising anti-aging in-
terventions in large clinical trials.
CoQ10 ameliorates some aging-related pathological processes and
may improve survival in patients with heart failure (Alehagen et al.,
2018;Lehagen et al., 2018;Li and Yan, 2017;Zhai et al., 2017).
Therefore, it might work as an anti-aging drug, although more research
in people without heart failure is needed.
Some antidiabetic drugs, such as metformin (Castro Cabezas et al.,
2012;de Kreutzenberg et al., 2015;Domecq et al., 2015;Haukka et al.,
2017;Wu et al., 2015a), GLP-1 agonists (Fatima et al., 2017) and
SGLT2 inhibitors (Bekki et al., 2018;Fatima et al., 2017;Shigiyama
et al., 2017) provide anti-aging benets and extend lifespan and
healthspan in people with diabetes and are tested for other conditions.
RAAS blocking drugs (Kim et al., 2016b;Wu et al., 2015a) act as anti-
aging interventions in patients with hypertension, but they are also
used against brotic diseases.
Statin use addresses some (but not all) pathological processes of
aging and reduces all-cause mortality, at least in some groups of people
(e.g. those with dyslipidemia) (Boccardi et al., 2013;Chou et al., 2014,
2016;Geifman et al., 2017;Kim et al., 2017;Li et al., 2017a;Naci et al.,
2013;Paradisi et al., 2012;Riekse et al., 2006;Smith et al., 2017).
The use of bisphosphonates improves survival in cancer patients
with benets generally outweighing the risks (Van Acker et al., 2016),
and may reduce all-cause mortality in patients with osteoporosis
(Kranenburg et al., 2016) although this nding was challenges recently
(Cummings et al., 2019).
Pentoxifylline (a PDE-3 inhibitor) may have anti-aging properties
(Champion et al., 2014;Chen et al., 2017c;McCarty et al., 2016;Pollice
et al., 2001;Wen et al., 2017), but more clinical trials on dierent in-
dications are needed. Interestingly, PDE-5 inhibitors in addition to
treating erectile dysfunction might have anti-aging eects in people
with a number of aging-related conditions like T2D (Anderson et al.,
2016) and heart failure (De Vecchis et al., 2017).
Glucosamine and chondroitin use also seems to act as an anti-aging
compound in its users (people with osteoarthritis) (Bell et al., 2012;
Katoh et al., 2017;Navarro et al., 2015).
Other promising but less studied interventions include N-acet-
ylcysteine (Ahmadi et al., 2017;Chughlay et al., 2016;Firuzi et al.,
2011;Talasaz et al., 2014), carnosine and close compounds
(Babizhayev et al., 2012;Babizhayev and Yegorov, 2016;de Courten
et al., 2016;Hisatsune et al., 2016;Menon et al., 2018;Regazzoni et al.,
2016), melatonin (Akbari et al., 2018;Doosti-Irani et al., 2018;
Hardeland et al., 2015;Raygan et al., 2017;Seely et al., 2012), NAD-
inducing substances including acipimox (Airhart et al., 2017;Trammell
et al., 2016;van de Weijer et al., 2015;Yoshino et al., 2018), probiotics
(Kasińska and Drzewoski, 2015;Lei et al., 2017;Wu et al., 2017;Zhang
et al., 2015), as well as plant-based quercetin (Askari et al., 2012;Byun
et al., 2017;Justice et al., 2019;Khan et al., 2016;Mohammadi-Sartang
et al., 2017;Nenna et al., 2015;Nieman et al., 2010;Sahebkar, 2017;
Serban et al., 2016), resveratrol (Chilelli et al., 2016;Guo et al., 2017;
Mousavi et al., 2018b;Tabrizi et al., 2018a;Zhao et al., 2019a), ber-
berine (Lan et al., 2015a), ginger (Maharlouei et al., 2018;Mazidi et al.,
2016), turmeric and curcumin (Chilelli et al., 2016;Mousavi et al.,
2018a;Tabrizi et al., 2018b), astragalus (Dow and Harley, 2016;He
et al., 2013;Tian et al., 2016;Wu et al., 2009), and olive-based pro-
ducts (de Bock et al., 2013;Fabiani, 2016;Filip et al., 2015;Lockyer
et al., 2017;Menicacci et al., 2017).
Certain regimes of HRT for post-menopausal women and men with
hypogonadism might have anti-aging eects (Lobo, 2017;Marjoribanks
et al., 2017), but more research is needed. Bariatric surgeries may
prevent pathologies of aging in people with obesity (Cardoso et al.,
2017).
This list is obviously incomplete, and it can be completed in the
future as geroprotectors identied in animal models will be tested in
humans (Moskalev et al., 2017), as well as with new clinical studies of
other interventions. So far we can generally conclude that the rate of
human aging does respond to interventions, both in general population
and patients with certain diseases.
3.5. Linkage to genetic factors: e.g., genotypes, patterns of gene expression
So far, only a few gene variants consistently associated with long-
evity in dierent ethnic groups have been identied (Dato et al., 2019).
Many of them are related to aging-related pathological processes, in-
cluding APOE (proteostasis, lipid metabolism) (Deelen et al., 2019),
FOXO3A, G vs T alleles, rs2802292 (defense against oxidative stress,
telomere length (Davy et al., 2018), cell survival, cardiovascular health,
etc.), KLOTHO KL-VS (calcium homeostasis, insulin sensitivity)
(Revelas et al., 2018), CDKN2A/B (cellular senescence) (Deelen et al.,
2019), IL-6 (inammation) (Deelen et al., 2019), and rs2149954 on
chromosome 5q33.3 (cardiovascular health) (Nygaard et al., 2017). The
role of other longevity-associated gene variants remains to be fully
understood, i.e. rs7676745, located on chromosome 4 near GPR78
(Deelen et al., 2019). Interestingly, growth hormone receptor exon 3
deletion is associated with longevity in men (Ben-Avraham et al.,
2017).
Studies in some specic groups reveal other interesting longevity
gene candidates potentially relevant for new therapy development.
Thus, a study of longevity among Amish community identied that
carriers of the null SERPINE1 allele had a longer lifespan (Khan et al.,
2017). Studies in California identied a relationship between longevity
and dopamine D4 receptor DRD4 gene (Grady et al., 2013;Szekely
et al., 2016) which may be due to neural system metabolism in old age
(Volkow et al., 2013).
Genetic studies of the multi-ethnic oldest-old populations reveal the
importance of DNA repair and proteostasis mechanisms which may be
encoded by dierent gene variants (Kim et al., 2018b).
Hopefully, further genetic research will identify more gene variants
related to longevity which will help to identify pro-longevity clinical
strategies.
Aging is associated with signicant aging-related epigenetic
changes (de Magalhães et al., 2009;Gopalan et al., 2017;Jung et al.,
2019;Li et al., 2019;Pal and Tyler, 2016;Salas-Pérez et al., 2019), but
their clinical signicance, as discussed earlier, remains currently to be
fully understood.
3.6. Linkage to interacting environmental factor
Environmental factors have large-scale eects on the rate of aging.
In fact, a safer environment with better food security and pathogen
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
14
control has allowed modern humans to achieve higher life expectancy
levels than ever before.
A systematic review of behavioral and metabolic risk factors of
disability-adjusted years of life lost identied poor control of high
systolic blood pressure, high fasting plasma glucose, smoking and high
BMI as major risk factors globally, each of them being a signicant
factor of accelerated aging as discussed above (GBD 2017 Risk Factor
Collaborators, 2018). In fact, smoking is a major risk factor of aging-
associated diseases, all-cause mortality (Mons et al., 2015) and ac-
celerated aging (Mamoshina et al., 2019).
Socioeconomic status is associated with higher life expectancy
(Stringhini et al., 2017). Also, education is negatively associated with
the risks of non-communicable diseases other than neoplastic diseases
(Smith et al., 2015).
Education, smoking, BMI, alcohol intake, and physical activity are
correlated with biomarkers of epigenetic aging (Fiorito et al., 2019).
Certain work conditions, such as night-shift work (Lin et al., 2015),
strenuous occupational physical activity (Coenen et al., 2018) or pro-
longed occupational sitting (Kikuchi et al., 2015) might impede health
and accelerate aging.
Environmental toxic pollution is associated with increased mortality
from aging-related diseases (Chowdhury et al., 2018;Pope et al., 2019).
Social engagement (Rico-Uribe et al., 2018), meaningful work, and
volunteering (Anderson et al., 2014;Rogers et al., 2016) are associated
with improved survival.
Characteristics of a neighborhood and built-in environment may
aect health in older ages (Malambo et al., 2016). A highly-walkable or
mountain environment predisposes individuals to physical activity and
longevity (Pes et al., 2013).
Therefore, aging can be attenuated, and healthy longevity main-
tained via various modications of the environment through the life-
course.
4. Discussion
Our review is obviously not comprehensive and should be further
developed. However, it does represent the rst systematic approach to
implementing aging in the ICD. Using the WHOs criteria for disease
classication, we conclude that aging generally ts all of them, and
therefore can be considered a disease.
We identied pathogenetic processes common for aging of dierent
organs and systems of the human body, according to the current state of
medical research. The major ones include aging-related low-grade
systemic inammation, replicative cellular senescence, proteostasis
failures, immunosenescence, mitochondrial dysfunctions, increased -
brotic propensity, insulin resistance, body composition changes and
hormonal changes in aging. Aging of some human body systems and
organs, including cardiovascular, gastroenterological, neural and
kidney aging, leads to the rise of secondary pathogenetic cascades of
aging which can harm the aging organism as a whole. These identied
pathogenetic processes aect many human body organs and systems.
We mapped connections between various pathogenetic processes of
aging which were described in the relevant sections of this study above
and summarized them in Fig. 1. The analysis of pathogenetic me-
chanisms of aging shows their high degree of interconnection, which
presents aging as a complex but a holistic process. Immune senescence
and some aspects of hormonal aging stand out to some extent as less
connected to other components of aging, which makes them less sus-
ceptible to therapeutic interventions.
Unlike some previous systematic studies of aging (de Grey and Rae,
2007;López-Otín et al., 2013), our review was almost completely
conned to human studies. Our results are rather similar to these au-
thors, which reects growing understanding and consensus on the
mechanisms of aging within the scientic community. Interestingly, our
study has identied aging-related increased brotic propensity as an as-
yet unrecognized hallmark of aging, in addition to traditionally
suspected ones. We would also like to stress the importance of further
human studies of epigenetic aspects of aging, DNA damage, as well as
calcium metabolism in aging. Some important aspects of aging were not
well studied in detail in human studies and could be added to research
reviews on human aging in the future. We hope that future research on
aging will allow for the development of a more unifying framework that
can reduce fragmentation of the various approaches, indicators and
interventions.
A number of interventions were identied, including lifestyle
changes, approved drugs and supplements, which proved eective for
the prevention of more than one aging-related pathological process.
We feel that our success in introducing aging into the ICD is an
important step toward opening the doors to treating aging-related dis-
eases. However, the need to include aging per se as a unitary disease or
a separate classication of aging-related pathologies in the future ICD
frameworks is still present in order to facilitate the development of
clinical practice guidelines and therapies to alleviate health burden
associated with aging and aging-related diseases (Calimport, 2019).
5. Conclusions
This review shows that human aging can be described in terms of
the WHO criteria for disease, including symptomatology, aetiology,
course and outcomes of the disease, possible and potential interven-
tions, and linkage to genetic and environmental factors. Pathogenetic
processes of aging are highly interconnected (Fig. 1), and therefore,
aging can be dened as complex of several mutually-inuencing pa-
thological processes.
The set of major pathogenetic processes of aging identied in this
study can be used for the development of clinically signicant panels of
aging biomarkers targeting individual pathological processes.
Finally, we conclude that aging ts all of the six disease criteria
developed for the ICD-11 and can be considered a disease. The argu-
ment whether aging is a disease or a syndrome may look like a merely
semantic matter, however, considering aging a disease may accelerate
the development and implementation of innovative technologies to
prevent and cure aging-related pathologies.
Declaration of Competing Interest
There is no conict of interest to declare.
Acknowledgments
We thank Ivan Alexeev, Edouard Debonneuil, John Furber, Aubrey
de Grey, Steven Hill, Boris Kaurov, Valentina Matveyeva, Aleksey
Moskalev, Valeria Pride, Paul Spiegel, Valeria Udalova, Alex
Zhavoronkov, Danila Medvedev, Arkadi Mazin and Viktor Zykov for the
assistance and advice.
Appendix A. Supplementary data
Supplementary material related to this article can be found, in the
online version, at doi:https://doi.org/10.1016/j.mad.2020.111230.
References
Abdelaal, M., le Roux, C.W., Docherty, N.G., 2017. Morbidity and mortality associated
with obesity. Annals of Translational Medicine 5. https://doi.org/10.21037/atm.
2017.03.107.
Aboyans, V., Ricco, J.-B., Bartelink, M.-L.E.L., Bjorck, M., Brodmann, M., Cohnert, T.,
Collet, J.-P., Czerny, M., De Carlo, M., Debus, S., 2018. 2017 ESC Guidelines on the
Diagnosis and Treatment of Peripheral Arterial Diseases, in collaboration with the
European Society for Vascular Surgery (ESVS): Document covering atherosclerotic
disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower
extremity arteries Endorsed by: the European Stroke Organization (ESO)The Task
Force for the Diagnosis and Treatment of Peripheral Arterial Diseases of the European
Society of Cardiology (ESC) and of the European Society for Vascular Surgery (ESVS).
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
15
European Heart Journal 39, 763816.
Ahmadi, F., Abbaszadeh, M., Razeghi, E., Maziar, S., Khoidaki, S.D., Naja, M.T., Lessan-
Pezeshki, M., 2017. Eectiveness of N-acetylcysteine for preserving residual renal
function in patients undergoing maintenance hemodialysis: multicenter randomized
clinical trial. Clinical and experimental nephrology 21, 342349.
Airhart, S.E., Shireman, L.M., Risler, L.J., Anderson, G.D., Gowda, G.A.N., Raftery, D.,
Tian, R., Shen, D.D., OBrien, K.D., 2017. An open-label, non-randomized study of the
pharmacokinetics of the nutritional supplement nicotinamide riboside (NR) and its
eects on blood NAD+ levels in healthy volunteers. PLoS One 12, e0186459.
Akasheva, D.U., Plokhova, E.V., Tkacheva, O.N., Strazhesko, I.D., Dudinskaya, E.N.,
Kruglikova, A.S., Pykhtina, V.S., Brailova, N.V., Pokshubina, I.A., Sharashkina, N.V.,
Agaltsov, M.V., Skvortsov, D., Boytsov, S.A., 2015. Age-Related Left Ventricular
Changes and Their Association with Leukocyte Telomere Length in Healthy People.
PLoS One 10, e0135883.
Akbari, M., Ostadmohammadi, V., Tabrizi, R., Lankarani, K.B., Heydari, S.T., Amirani, E.,
Reiter, R.J., Asemi, Z., 2018. The eects of melatonin supplementation on in-
ammatory markers among patients with metabolic syndrome or related disorders: a
systematic review and meta-analysis of randomized controlled trials.
Inammopharmacology 26, 899907.
Alehagen, U., Aaseth, J., Alexander, J., Johansson, P., 2018. Still reduced cardiovascular
mortality 12 years after supplementation with selenium and coenzyme Q10 for four
years: A validation of previous 10-year follow-up results of a prospective randomized
double-blind placebo-controlled trial in elderly. PLoS One 13, e0193120.
Alfaris, N., Minnick, A.M., Hopkins, C.M., Berkowitz, R.I., Wadden, T.A., 2015.
Combination phentermine and topiramate extended release in the management of
obesity. Expert Opin. Pharmacother. 16, 12631274.
Alique, M., Ramírez-Carracedo, R., Bodega, G., Carracedo, J., Ramírez, R., 2018.
Senescent Microvesicles: A Novel Advance in Molecular Mechanisms of
Atherosclerotic Calcication. Int. J. Mol. Sci. 19. https://doi.org/10.3390/
ijms19072003.
Alves, A.J., Viana, J.L., Cavalcante, S.L., Oliveira, N.L., Duarte, J.A., Mota, J., Oliveira, J.,
Ribeiro, F., 2016. Physical activity in primary and secondary prevention of cardio-
vascular disease: Overview updated. World J. Cardiol. 8, 575.
Ambrose, C.T., 2017. Pro-Angiogenesis Therapy and Aging: A Mini-Review. Gerontology
63, 393400.
Amir, M., Czaja, M.J., 2011. Autophagy in nonalcoholic steatohepatitis. Expert Rev.
Gastroenterol. Hepatol. 5, 159166.
Andersen, S.L., Sebastiani, P., Dworkis, D.A., Feldman, L., Perls, T.T., 2012. Health span
approximates life span among many supercentenarians: compression of morbidity at
the approximate limit of life span. J. Gerontol. A Biol. Sci. Med. Sci. 67, 395405.
Anderson, N.D., Damianakis, T., Kroger, E., Wagner, L.M., Dawson, D.R., Binns, M.A.,
Bernstein, S., Caspi, E., Cook, S.L., 2014. The benets associated with volunteering
among seniors: a critical review and recommendations for future research.
Psychological bulletin 140, 15051533.
Anderson, S.G., Hutchings, D.C., Woodward, M., Rahimi, K., Rutter, M.K., Kirby, M.,
Hackett, G., Traord, A.W., Heald, A.H., 2016. Phosphodiesterase type-5 inhibitor
use in type 2 diabetes is associated with a reduction in all-cause mortality. Heart 102,
1750.
Andersson, D.P., Lagerros, Y.T., Grotta, A., Bellocco, R., Lehtihet, M., Holzmann, M.J.,
2017. Association between treatment for erectile dysfunction and death or cardio-
vascular outcomes after myocardial infarction. Heart 103, 12641270.
Araujo, A.B., Dixon, J.M., Suarez, E.A., Murad, M.H., Guey, L.T., Wittert, G.A., 2011.
Clinical review: Endogenous testosterone and mortality in men: a systematic review
and meta-analysis. J. Clin. Endocrinol. Metab. 96, 30073019.
Arem, H., Moore, S.C., Patel, A., Hartge, P., Berrington de Gonzalez, A., Visvanathan, K.,
Campbell, P.T., Freedman, M., Weiderpass, E., Adami, H.O., Linet, M.S., Lee, I.-M.,
Matthews, C.E., 2015. Leisure time physical activity and mortality: a detailed pooled
analysis of the dose-response relationship. JAMA Intern. Med. 175, 959967.
Askari, G., Ghiasvand, R., Feizi, A., Ghanadian, S.M., Karimian, J., 2012. The eect of
quercetin supplementation on selected markers of inammation and oxidative stress.
J. Res. Med. Sci. 17, 637641.
Austad, S.N., Fischer, K.E., 2016. Sex Dierences in Lifespan. Cell Metab. 23, 10221033.
Ayaki, M., Shiba, D., Negishi, K., Tsubota, K., 2016. Depressed visual eld and mood are
associated with sleep disorder in glaucoma patients. Sci. Rep. 6, 25699.
Babizhayev, M.A., Deyev, A.I., SavelYeva, E.L., Lankin, V.Z., Yegorov, Y.E., 2012. Skin
beautication with oral non-hydrolized versions of carnosine and carcinine: Eective
therapeutic management and cosmetic skincare solutions against oxidative glycation
and free-radical production as a causal mechanism of diabetic complications and skin
aging. Journal of Dermatological Treatment 23, 345384.
Babizhayev, M.A., Yegorov, Y.E., 2016. Telomere Attrition in Human Lens Epithelial Cells
Associated with Oxidative Stress Provide a New Therapeutic Target for the
Treatment, Dissolving and Prevention of Cataract with N-Acetylcarnosine Lubricant
Eye Drops. Kinetic, Pharmacological and Activity-Dependent Separation of
Therapeutic Targeting: Transcorneal Penetration and Delivery of L-Carnosine in the
Aqueous Humor and Hormone-Like Hypothalamic Antiaging Eects of the Instilled
Ophthalmic Drug Through a Safe Eye Medication Technique. Recent Pat. Drug Deliv.
Formul. 10, 82129.
Baker, L.D., Sambamurti, K., Craft, S., Cherrier, M., Raskind, M.A., Stanczyk, F.Z.,
Plymate, S.R., Asthana, S., 2003. 17beta-estradiol reduces plasma Abeta40 for HRT-
naïve postmenopausal women with Alzheimer disease: a preliminary study. Am. J.
Geriatr. Psychiatry 11, 239244.
Bann, D., Wu, F.C.W., Keevil, B., Lashen, H., Adams, J., Hardy, R., Muniz, G., Kuh, D.,
Ben-Shlomo, Y., Ong, K.K., 2015. Changes in testosterone related to body composi-
tion in late midlife: Findings from the 1946 British birth cohort study. Obesity 23,
14861492.
Barron, A.M., Pike, C.J., 2012. Sex hormones, aging, and Alzheimers disease. Front.
Biosci. 4, 976997.
Barron, E., Lara, J., White, M., Mathers, J.C., 2015. Blood-borne biomarkers of mortality
risk: systematic review of cohort studies. PLoS One 10, e0127550.
Bartolome, F., de la Cueva, M., Pascual, C., Antequera, D., Fernandez, T., Gil, C.,
Martinez, A., Carro, E., 2018. Amyloid β-induced impairments on mitochondrial
dynamics, hippocampal neurogenesis, and memory are restored by phosphodies-
terase 7 inhibition. Alzheimers. Res. Ther. 10, 24.
Baud, S., Duca, L., Bochicchio, B., Brassart, B., Belloy, N., Pepe, A., Dauchez, M., Martiny,
L., Debelle, L., 2013. Elastin peptides in aging and pathological conditions. Biomol.
Concepts 4, 6576.
Baykal, Y., Yilmaz, M.I., Celik, T., Gok, F., Rehber, H., Akay, C., Kocar, I.H., 2003. Eects
of antihypertensive agents, alpha receptor blockers, beta blockers, angiotensin-con-
verting enzyme inhibitors, angiotensin receptor blockers and calcium channel
blockers, on oxidative stress. J. Hypertens. 21, 12071211.
Bays, H.E., Gadde, K.M., 2011. Phentermine/topiramate for weight reduction and treat-
ment of adverse metabolic consequences in obesity. Drugs Today 47, 903914.
Bechthold, A., Boeing, H., Schwedhelm, C., Homann, G., Knüppel, S., Iqbal, K., De
Henauw, S., Michels, N., Devleesschauwer, B., Schlesinger, S., Schwingshackl, L.,
2017. Food groups and risk of coronary heart disease, stroke and heart failure: A
systematic review and dose-response meta-analysis of prospective studies. Crit. Rev.
Food Sci. Nutr. 120.
Beerman, I., Seita, J., Inlay, M.A., Weissman, I.L., Rossi, D.J., 2014. Quiescent hemato-
poietic stem cells accumulate DNA damage during aging that is repaired upon entry
into cell cycle. Cell Stem Cell 15, 37.
Bekki, M., Tahara, N., Tahara, A., Igata, S., Honda, A., Sugiyama, Y., Nakamura, T., Sun,
J., Kumashiro, Y., Matsui, T., Fukumoto, Y., Yamagishi, S.-I., 2018. Switching di-
peptidyl peptidase-4 inhibitors to tofogliozin, a selective inhibitor of sodium-glu-
cose cotransporter 2 improves arterial stiness evaluated by cardio-ankle vascular
index in patients with type 2 diabetes: a pilot study. Curr. Vasc. Pharmacol. https://
doi.org/10.2174/1570161116666180515154555.
Bella, F., Godos, J., Ippolito, A., Di Prima, A., Sciacca, S., 2017. Dierences in the asso-
ciation between empirically derived dietary patterns and cancer: a meta-analysis. Int.
J. Food Sci. Nutr. 68, 402410.
Bell, G.A., Kantor, E.D., Lampe, J.W., Shen, D.D., White, E., 2012. Use of glucosamine and
chondroitin in relation to mortality. Eur. J. Epidemiol. 27, 593603.
Bell, K.E., Snijders, T., Zulyniak, M., Kumbhare, D., Parise, G., Chabowski, A., Phillips,
S.M., 2017. A whey protein-based multi-ingredient nutritional supplement stimulates
gains in lean body mass and strength in healthy older men: A randomized controlled
trial. PLoS One 12, e0181387.
Bellon, M., Nicot, C., 2017. Telomere Dynamics in Immune Senescence and Exhaustion
Triggered by Chronic Viral Infection. Viruses 9. https://doi.org/10.3390/v9100289.
Belsky, D.W., Caspi, A., Houts, R., Cohen, H.J., Corcoran, D.L., Danese, A., Harrington, H.,
Israel, S., Levine, M.E., Schaefer, J.D., Sugden, K., Williams, B., Yashin, A.I., Poulton,
R., Mott, T.E., 2015. Quantication of biological aging in young adults.
Proceedings of the National Academy of Sciences 112, E4104E4110.
Benatti, F.B., Ried-Larsen, M., 2015. The Eects of Breaking up Prolonged Sitting Time: A
Review of Experimental Studies. Med. Sci. Sports Exerc. 47, 20532061.
Ben-Avraham, D., Govindaraju, D.R., Budagov, T., Fradin, D., Durda, P., Liu, B., Ott, S.,
Gutman, D., Sharvit, L., Kaplan, R., Bougnères, P., Reiner, A., Shuldiner, A.R., Cohen,
P., Barzilai, N., Atzmon, G., 2017. The GH receptor exon 3 deletion is a marker of
male-specic exceptional longevity associated with increased GH sensitivity and
taller stature. Sci Adv 3, e1602025.
Benilova, I., Karran, E., De Strooper, B., 2012. The toxic Aβoligomer and Alzheimers
disease: an emperor in need of clothes. Nat. Neurosci. 15, 349357.
Bernstein, A.M., Willcox, B.J., Tamaki, H., Kunishima, N., Suzuki, M., Willcox, D.C., Yoo,
J.-S.K., Perls, T.T., 2004. First autopsy study of an Okinawan centenarian: absence of
many age-related diseases. J. Gerontol. A Biol. Sci. Med. Sci. 59, 11951199.
Beswick, M., Pachnio, A., Lauder, S.N., Sweet, C., Moss, P.A., 2013. Antiviral therapy can
reverse the development of immune senescence in elderly mice with latent cytome-
galovirus infection. J. Virol. 87, 779789.
Bhat, R., Crowe, E.P., Bitto, A., Moh, M., Katsetos, C.D., Garcia, F.U., Johnson, F.B.,
Trojanowski, J.Q., Sell, C., Torres, C., 2012. Astrocyte senescence as a component of
Alzheimers disease. PLoS One 7, e45069.
Bianchi, E., Scarinci, F., Ripandelli, G., Feher, J., Pacella, E., Magliulo, G., Gabrieli, C.B.,
Plateroti, R., Plateroti, P., Mignini, F., Artico, M., 2013. Retinal pigment epithelium,
age-related macular degeneration and neurotrophic keratouveitis. Int. J. Mol. Med.
31, 232242.
Blaise, S., Romier, B., Kawecki, C., Ghirardi, M., Rabenoelina, F., Baud, S., Duca, L.,
Maurice, P., Heinz, A., Schmelzer, C.E.H., Tarpin, M., Martiny, L., Garbar, C.,
Dauchez, M., Debelle, L., Durlach, V., 2013. Elastin-Derived Peptides Are New
Regulators of Insulin Resistance Development in Mice. Diabetes 62, 3807.
Boada, M., Anaya, F., Ortiz, P., Olazarán, J., Shua-Haim, J.R., Obisesan, T.O., Hernández,
I., Muñoz, J., Buendia, M., Alegret, M., Lafuente, A., Tárraga, L., Núñez, L., Torres,
M., Grifols, J.R., Ferrer, I., Lopez, O.L., Páez, A., 2017. Ecacy and Safety of Plasma
Exchange with 5% Albumin to Modify Cerebrospinal Fluid and Plasma Amyloid-β
Concentrations and Cognition Outcomes in Alzheimers Disease Patients: A
Multicenter, Randomized, Controlled Clinical Trial. J. Alzheimers. Dis. 56, 129.
Boccardi, V., Barbieri, M., Rizzo, M.R., Marfella, R., Esposito, A., Marano, L., Paolisso, G.,
2013. A new pleiotropic eect of statins in elderly: modulation of telomerase activity.
FASEB J. 27, 38793885.
Bouajila, S., Moneghetti, K.J., Kobayashi, Y., Abbasi, F., Gomari-Grisar, F.A., Kuznetsova,
T., Wu, J.C., Haddad, F., 2018. Biomarker and cardiovascular proles of healthy
aging, putting multiple biomarkers in perspective. Archives of Cardiovascular
Diseases Supplements 10, 107108.
Bouitbir, J., Charles, A.-L., Echaniz-Laguna, A., Kindo, M., Daussin, F., Auwerx, J.,
Piquard, F., Geny, B., Zoll, J., 2012. Opposite eects of statins on mitochondria of
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
16
cardiac and skeletal muscles: a mitohormesismechanism involving reactive oxygen
species and PGC-1. Eur. Heart J. 33, 13971407.
Brandenberger, C., Mühlfeld, C., 2017. Mechanisms of lung aging. Cell Tissue Res. 367,
469480.
Brasky, T.M., Lampe, J.W., Slatore, C.G., White, E., 2011. Use of glucosamine and
chondroitin and lung cancer risk in the VITamins And Lifestyle (VITAL) cohort.
Cancer Causes Control 22, 13331342.
Brehme, M., Voisine, C., Rolland, T., Wachi, S., Soper, J.H., Zhu, Y., Orton, K., Villella, A.,
Garza, D., Vidal, M., Ge, H., Morimoto, R.I., 2014. A Chaperome Sub-Network
Safeguards Proteostasis in Aging and Neurodegenerative Disease. Cell Rep. 9, 1135.
Brierley, E.J., Johnson, M.A., Lightowlers, R.N., James, O.F., Turnbull, D.M., 1998. Role
of mitochondrial DNA mutations in human aging: implications for the central nervous
system and muscle. Ann. Neurol. 43, 217223.
Briley, S.M., Jasti, S., McCracken, J.M., Hornick, J.E., Fegley, B., Pritchard, M.T., Duncan,
F.E., 2016. Reproductive age-associated brosis in the stroma of the mammalian
ovary. Reproduction 152, 245260.
Brooks, W.M., Lynch, P.J., Ingle, C.C., Hatton, A., Emson, P.C., Faull, R.L.M., Starkey,
M.P., 2007. Gene expression proles of metabolic enzyme transcripts in Alzheimers
disease. Brain Res. 1127, 127135.
Bua, E., Johnson, J., Herbst, A., Delong, B., McKenzie, D., Salamat, S., Aiken, J.M., 2006.
Mitochondrial DNADeletion Mutations Accumulate Intracellularly to Detrimental
Levels in Aged Human Skeletal Muscle Fibers. Am. J. Hum. Genet. 79, 469480.
Buckinx, F., Rolland, Y., Reginster, J.-Y., Ricour, C., Petermans, J., Bruyère, O., 2015.
Burden of frailty in the elderly population: perspectives for a public health challenge.
Arch. Public Health 73, 19.
Bulterijs, S., Hull, R.S., Björk, V.C.E., Roy, A.G., 2015. It is time to classify biological aging
as a disease. Front. Genet. 6, 205.
Bürkle, A., Moreno-Villanueva, M., Bernhard, J., Blasco, M., Zondag, G., Hoeijmakers,
J.H.J., Toussaint, O., Grubeck-Loebenstein, B., Mocchegiani, E., Collino, S., Gonos,
E.S., Sikora, E., Gradinaru, D., Dollé, M., Salmon, M., Kristensen, P., Griths, H.R.,
Libert, C., Grune, T., Breusing, N., Simm, A., Franceschi, C., Capri, M., Talbot, D.,
Caiafa, P., Friguet, B., Slagboom, P.E., Hervonen, A., Hurme, M., Aspinall, R., 2015.
MARK-AGE biomarkers of ageing. Mech. Ageing Dev. 151, 212.
Burtscher, M., 2013. Exercise limitations by the oxygen delivery and utilization systems in
aging and disease: coordinated adaptation and deadaptation of the lung-heart muscle
axis - a mini-review. Gerontology 59, 289296.
Byun, K., Yoo, Y., Son, M., Lee, J., Jeong, G.-B., Park, Y.M., Salekdeh, G.H., Lee, B., 2017.
Advanced glycation end-products produced systemically and by macrophages: A
common contributor to inammation and degenerative diseases. Pharmacol. Ther.
177, 4455.
Calhoun, C., Shivshankar, P., Saker, M., Sloane, L.B., Livi, C.B., Sharp, Z.D., Orihuela,
C.J., Adnot, S., White, E.S., Richardson, A., Le Saux, C.J., 2016. Senescent Cells
Contribute to the Physiological Remodeling of Aged Lungs. J. Gerontol. A Biol. Sci.
Med. Sci. 71, 153.
Calimport, S., et al., 2019. To help aging populations, classify organismal senescence.
Science 1 (366), 576578. https://doi.org/10.1126/science.aay7319.
Calimport, S., Bentley, B., 2019. Ageing classied as a cause of disease in ICD11.
Rejuvenation Res. 22 (4), 281. https://doi.org/10.1089/rej.2019.2242.
Camões, J., Coelho, A., Castro-Diaz, D., Cruz, F., 2015. Lower Urinary Tract Symptoms
and Aging: The Impact of Chronic Bladder Ischemia on Overactive Bladder
Syndrome. Urol. Int. 95, 373379.
Campbell, J.M., Bellman, S.M., Stephenson, M.D., Lisy, K., 2017. Metformin reduces all-
cause mortality and diseases of ageing independent of its eect on diabetes control: a
systematic review and meta-analysis. Ageing Res. Rev. 40, 3144.
Campisi, J., 2013. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75,
685705.
Cardoso, L., Rodrigues, D., Gomes, L., Carrilho, F., 2017. Short- and long-term mortality
after bariatric surgery: A systematic review and meta-analysis. Diabetes Obes. Metab.
19, 12231232.
Carreira, H., Pereira, M., Alves, L., Lunet, N., Azevedo, A., 2012. Dyslipidaemia, and mean
blood cholesterol and triglycerides levels in the Portuguese population: a systematic
review. Arq. Med. 26, 112123.
Castro Cabezas, M., van Wijk, J.P.H., Elte, J.W.F., Klop, B., 2012. Eects of metformin on
the regulation of free Fatty acids in insulin resistance: a double-blind, placebo-con-
trolled study. J. Nutr. Metab., 394623 2012.
Cawood, A.L., Elia, M., Stratton, R.J., 2012. Systematic review and meta-analysis of the
eects of high protein oral nutritional supplements. Ageing Res. Rev. 11, 278296.
Cawthon, R.M., Smith, K.R., OBrien, E., Sivatchenko, A., Kerber, R.A., 2003. Association
between telomere length in blood and mortality in people aged 60 years or older.
Lancet 361, 393395393395.
Cesari, M., Fielding, R., Bénichou, O., Bernabei, R., Bhasin, S., Guralnik, J.M., Jette, A.,
Landi, F., Pahor, M., Rodriguez-Manas, L., Rolland, Y., Roubeno, R., Sinclair, A.J.,
Studenski, S., Travison, T., Vellas, B., 2015. Pharmacological interventions in frailty
and sarcopenia: report by the international conference on frailty and sarcopenia re-
search task force. J Frailty Aging 4, 114120.
Champion, S., Lapidus, N., Cherié, G., Spagnoli, V., Oliary, J., Solal, A.C., 2014.
Pentoxifylline in heart failure: a meta-analysis of clinical trials. Cardiovasc. Ther. 32,
159162.
Chan, D.T., Watts, G.F., Irish, A.B., Dogra, G.K., 2017. Insulin resistance and vascular
dysfunction in chronic kidney disease: mechanisms and therapeutic interventions.
Nephrol. Dial. Transplant 32, 12741281.
Chaudhary, N.I., Roth, G.J., Hilberg, F., Müller-Quernheim, J., Prasse, A., Zissel, G.,
Schnapp, A., Park, J.E., 2007. Inhibition of PDGF, VEGF and FGF signalling attenu-
ates brosis. Eur. Respir. J. 29, 976985.
Chen, F., Liu, Y., Wong, N.-K., Xiao, J., So, K.-F., 2017a. Oxidative Stress in Stem Cell
Aging. Cell Transplant. 26, 1483.
Chen, G.-C., Zhang, R., Martínez-González, M.A., Zhang, Z.-L., Bonaccio, M., van Dam,
R.M., Qin, L.-Q., 2017b. Nut consumption in relation to all-cause and cause-specic
mortality: a meta-analysis 18 prospective studies. Food Funct. 8, 38933905.
Cheng, G., Huang, C., Deng, H., Wang, H., 2012. Diabetes as a risk factor for dementia and
mild cognitive impairment: a meta-analysis of longitudinal studies. Intern. Med. J.
42, 484491.
Chen, Y.-C., Hu, H.-Y., Fan, H.-Y., Kao, W.-S., Chen, H.-Y., Huang, S.-J., 2019. Where and
How Centenarians Die? The Role of Hospice Care. Am. J. Hosp. Palliat. Care 36,
10681075.
Chen, Y.-M., Chiang, W.-C., Lin, S.-L., Tsai, T.-J., 2017c. Therapeutic ecacy of pen-
toxifylline on proteinuria and renal progression: an update. J. Biomed. Sci. 24, 84.
Chilelli, N.C., Ragazzi, E., Valentini, R., Cosma, C., Ferraresso, S., Lapolla, A., Sartore, G.,
2016. Curcumin and Boswellia serrata Modulate the Glyco-Oxidative Status and Lipo-
Oxidation in Master Athletes. Nutrients 8. https://doi.org/10.3390/nu8110745.
Chini, E.N., Chini, C.C.S., Espindola Netto, J.M., de Oliveira, G.C., van Schooten, W.,
2018. The Pharmacology of CD38/NADase: An Emerging Target in Cancer and
Diseases of Aging. Trends Pharmacol. Sci. 39, 424436.
Chiti, F., Dobson, C.M., 2017. Protein Misfolding, Amyloid Formation, and Human
Disease: A Summary of Progress Over the Last Decade. Annu. Rev. Biochem. 86,
2768.
Chou, C.-Y., Chou, Y.-C., Chou, Y.-J., Yang, Y.-F., Huang, N., 2014. Statin use and incident
dementia: a nationwide cohort study of Taiwan. Int. J. Cardiol. 173, 305310.
Chou, R., Dana, T., Blazina, I., Daeges, M., Jeanne, T.L., 2016. Statins for Prevention of
Cardiovascular Disease in Adults: Evidence Report and Systematic Review for the US
Preventive Services Task Force. JAMA 316, 20082024.
Chowdhury, R., Kunutsor, S., Vitezova, A., Oliver-Williams, C., Chowdhury, S., Kiefte-de-
Jong, J.C., Khan, H., Baena, C.P., Prabhakaran, D., Hoshen, M.B., Feldman, B.S., Pan,
A., Johnson, L., Crowe, F., Hu, F.B., Franco, O.H., 2014. Vitamin D and risk of cause
specic death: systematic review and meta-analysis of observational cohort and
randomised intervention studies. BMJ 348, g1903.
Chowdhury, R., Ramond, A., OKeee, L.M., Shahzad, S., Kunutsor, S.K., Muka, T.,
Gregson, J., Willeit, P., Warnakula, S., Khan, H., Chowdhury, S., Gobin, R., Franco,
O.H., Di Angelantonio, E., 2018. Environmental toxic metal contaminants and risk of
cardiovascular disease: systematic review and meta-analysis. BMJ 362. https://doi.
org/10.1136/bmj.k3310.
Christou, M.A., Christou, P.A., Markozannes, G., Tsatsoulis, A., Mastorakos, G., Tigas, S.,
2017. Eects of Anabolic Androgenic Steroids on the Reproductive System of Athletes
and Recreational Users: A Systematic Review and Meta-Analysis. Sports Med. 47,
18691883.
Chughlay, M.F., Kramer, N., Spearman, C.W., Werfalli, M., Cohen, K., 2016. N-acet-
ylcysteine for non-paracetamol drug-induced liver injury: a systematic review. Br. J.
Clin. Pharmacol. 81, 10211029.
Cinti, S., Mitchell, G., Barbatelli, G., Murano, I., Ceresi, E., Faloia, E., Wang, S., Fortier,
M., Greenberg, A.S., Obin, M.S., 2005. Adipocyte death denes macrophage locali-
zation and function in adipose tissue of obese mice and humans. J. Lipid Res. 46,
23472355.
Cleasby, M.E., Jamieson, P.M., Atherton, P.J., 2016. Insulin resistance and sarcopenia:
mechanistic links between common co-morbidities. J. Endocrinol. 229, R6781.
Clement, J., Wong, M., Poljak, A., Sachdev, P., Braidy, N., 2018. The Plasma NAD +
Metabolome Is Dysregulated in NormalAging. Rejuvenation Res. https://doi.org/
10.1089/rej.2018.2077.
Coenen, P., Huysmans, M.A., Holtermann, A., Krause, N., van Mechelen, W., Straker,
L.M., van der Beek, A.J., 2018. Do highly physically active workers die early? A
systematic review with meta-analysis of data from 193 696 participants. Br. J. Sports
Med. 52, 13201326.
Coen, P.M., Jubrias, S.A., Distefano, G., Amati, F., Mackey, D.C., Glynn, N.W., Manini,
T.M., Wohlgemuth, S.E., Leeuwenburgh, C., Cummings, S.R., Newman, A.B.,
Ferrucci, L., Toledo, F.G.S., Shankland, E., Conley, K.E., Goodpaster, B.H., 2013.
Skeletal muscle mitochondrial energetics are associated with maximal aerobic ca-
pacity and walking speed in older adults. J. Gerontol. A Biol. Sci. Med. Sci. 68,
447455.
Cohen, A.A., Legault, V., Fuellen, G., Fülöp, T., Fried, L.P., Ferrucci, L., 2018. The risks of
biomarker-based epidemiology: Associations of circulating calcium levels with age,
mortality, and frailty vary substantially across populations. Exp. Gerontol. 107,
1117.
Conn, V.S., Koopman, R.J., Ruppar, T.M., Phillips, L.J., Mehr, D.R., Hafdahl, A.R., 2014.
Insulin Sensitivity Following Exercise Interventions: Systematic Review and Meta-
Analysis of Outcomes Among Healthy Adults. J. Prim. Care Community Health 5,
211222.
Coppé, J.-P., Desprez, P.-Y., Krtolica, A., Campisi, J., 2010. The senescence-associated
secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5,
99118.
Corona, G., Rastrelli, G., Giagulli, V.A., Sila, A., Sforza, A., Forti, G., Mannucci, E., Maggi,
M., 2013. Dehydroepiandrosterone Supplementation in Elderly Men: A Meta-Analysis
Study of Placebo-Controlled Trials. J. Clin. Endocrinol. Metab. 98, 36153626.
Crimmins, E., Vasunilashorn, S., Kim, J.K., Alley, D., 2008. Biomarkers related to aging in
human populations. Adv. Clin. Chem. 46, 161216.
Cuberas-Borrós, G., Roca, I., Boada, M., Tárraga, L., Hernández, I., Buendia, M., Rubio, L.,
Torres, G., Bittini, Á., Guzmán-de-Villoria, J.A., Pujadas, F., Torres, M., Núñez, L.,
Castell, J., Páez, A., 2018. Longitudinal Neuroimaging Analysis in Mild-Moderate
Alzheimers Disease Patients Treated with Plasma Exchange with 5% Human
Albumin. J. Alzheimers. Dis. 61, 321.
Cuccurullo, C., Iezzi, A., Fazia, M.L., De Cesare, D., Di Francesco, A., Muraro, R., Bei, R.,
Ucchino, S., Spigonardo, F., Chiarelli, F., Schmidt, A.M., Cuccurullo, F., Mezzetti, A.,
Cipollone, F., 2006. Suppression of RAGE as a basis of simvastatin-dependent plaque
stabilization in type 2 diabetes. Arterioscler. Thromb. Vasc. Biol. 26, 27162723.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
17
Cummings, S.R., Lui, L.-Y., Eastell, R., Allen, I.E., 2019. Association Between Drug
Treatments for Patients With Osteoporosis and Overall Mortality Rates: A Meta-
analysis. JAMA Intern. Med. https://doi.org/10.1001/jamainternmed.2019.2779.
Dai, D.-F., Chen, T., Johnson, S.C., Szeto, H., Rabinovitch, P.S., 2012. Cardiac Aging:
From Molecular Mechanisms to Signicance in Human Health and Disease. Antioxid.
Redox Signal. 16, 1492.
Darweesh, S.K., Ibrahim, M.F., El-Tahawy, M.A., 2017. Eect of N-Acetylcysteine on
Mortality and Liver Transplantation Rate in Non-Acetaminophen-Induced Acute Liver
Failure: A Multicenter Study. Clin. Drug Investig. 37, 473482.
Dato, S., Soerensen, M., Rose, G., 2019. Untangling the Genetics of Human Longevity-A
Challenging Quest. Genes 10. https://doi.org/10.3390/genes10080585.
Davies, M.J., Aronne, L.J., Caterson, I.D., Thomsen, A.B., Jacobsen, P.B., Marso, S.P.,
2018. Liraglutide and cardiovascular outcomes in adults with overweight or obesity:
A post hoc analysis from SCALE randomized controlled trials. Satiety and Clinical
Adiposity - Liraglutide Evidence in individuals with and without diabetes (SCALE)
study groups. Diabetes Obes. Metab. 20, 734739.
Davy, P.M.C., Willcox, D.C., Shimabukuro, M., Donlon, T.A., Torigoe, T., Suzuki, M.,
Higa, M., Masuzaki, H., Sata, M., Chen, R., Murkofsky, R.L., Morris, B.J., Lim, E.,
Allsopp, R.C., Willcox, B.J., 2018. Minimal Shortening of Leukocyte Telomere Length
Across Age Groups in a Cross-Sectional Study for Carriers of a Longevity-Associated
FOXO3 Allele. J. Gerontol. A Biol. Sci. Med. Sci. 73, 14481452.
Day, F.R., Ruth, K.S., Thompson, D.J., Lunetta, K.L., Pervjakova, N., Chasman, D.I., Stolk,
L., Finucane, H.K., Sulem, P., Bulik-Sullivan, B., Esko, T., Johnson, A.D., Elks, C.E.,
Franceschini, N., He, C., Altmaier, E., Brody, J.A., Franke, L.L., Human, J.E., Keller,
M.F., McArdle, P.F., Nutile, T., Porcu, E., Robino, A., Rose, L.M., Schick, U.M., Smith,
J.A., Teumer, A., Traglia, M., Vuckovic, D., Yao, J., Zhao, W., Albrecht, E., Amin, N.,
Corre, T., Hottenga, J.-J., Mangino, M., Smith, A.V., Tanaka, T., Abecasis, G.,
Andrulis, I.L., Anton-Culver, H., Antoniou, A.C., Arndt, V., Arnold, A.M., Barbieri, C.,
Beckmann, M.W., Beeghly-Fadiel, A., Benitez, J., Bernstein, L., Bielinski, S.J.,
Blomqvist, C., Boerwinkle, E., Bogdanova, N.V., Bojesen, S.E., Bolla, M.K., Borresen-
Dale, A.-L., Boutin, T.S., Brauch, H., Brenner, H., Brüning, T., Burwinkel, B.,
Campbell, A., Campbell, H., Chanock, S.J., Chapman, J.R., Chen, Y.-D.I., Chenevix-
Trench, G., Couch, F.J., Coviello, A.D., Cox, A., Czene, K., Darabi, H., De Vivo, I.,
Demerath, E.W., Dennis, J., Devilee, P., Dörk, T., Dos-Santos-Silva, I., Dunning, A.M.,
Eicher, J.D., Fasching, P.A., Faul, J.D., Figueroa, J., Flesch-Janys, D., Gandin, I.,
Garcia, M.E., García-Closas, M., Giles, G.G., Girotto, G.G., Goldberg, M.S., González-
Neira, A., Goodarzi, M.O., Grove, M.L., Gudbjartsson, D.F., Guénel, P., Guo, X.,
Haiman, C.A., Hall, P., Hamann, U., Henderson, B.E., Hocking, L.J., Hofman, A.,
Homuth, G., Hooning, M.J., Hopper, J.L., Hu, F.B., Huang, J., Humphreys, K., Hunter,
D.J., Jakubowska, A., Jones, S.E., Kabisch, M., Karasik, D., Knight, J.A., Kolcic, I.,
Kooperberg, C., Kosma, V.-M., Kriebel, J., Kristensen, V., Lambrechts, D., Langenberg,
C., Li, J., Li, X., Lindström, S., Liu, Y., Luan, J.an, Lubinski, J., Mägi, R., Mannermaa,
A., Manz, J., Margolin, S., Marten, J., Martin, N.G., Masciullo, C., Meindl, A.,
Michailidou, K., Mihailov, E., Milani, L., Milne, R.L., Müller-Nurasyid, M., Nalls, M.,
Neale, B.M., Nevanlinna, H., Neven, P., Newman, A.B., Nordestgaard, B.G., Olson,
J.E., Padmanabhan, S., Peterlongo, P., Peters, U., Petersmann, A., Peto, J., Pharoah,
P.D.P., Pirastu, N.N., Pirie, A., Pistis, G., Polasek, O., Porteous, D., Psaty, B.M., Pylkäs,
K., Radice, P., Rael, L.J., Rivadeneira, F., Rudan, I., Rudolph, A., Ruggiero, D., Sala,
C.F., Sanna, S., Sawyer, E.J., Schlessinger, D., Schmidt, M.K., Schmidt, F., Schmutzler,
R.K., Schoemaker, M.J., Scott, R.A., Seynaeve, C.M., Simard, J., Sorice, R., Southey,
M.C., Stöckl, D., Strauch, K., Swerdlow, A., Taylor, K.D., Thorsteinsdottir, U., Toland,
A.E., Tomlinson, I., Truong, T., Tryggvadottir, L., Turner, S.T., Vozzi, D., Wang, Q.,
Wellons, M., Willemsen, G., Wilson, J.F., Winqvist, R., Wolenbuttel, B.B.H.R.,
Wright, A.F., Yannoukakos, D., Zemunik, T., Zheng, W., Zygmunt, M., Bergmann, S.,
Boomsma, D.I., Buring, J.E., Ferrucci, L., Montgomery, G.W., Gudnason, V., Spector,
T.D., van Duijn, C.M., Alizadeh, B.Z., Ciullo, M., Crisponi, L., Easton, D.F., Gasparini,
P.P., Gieger, C., Harris, T.B., Hayward, C., Kardia, S.L.R., Kraft, P., McKnight, B.,
Metspalu, A., Morrison, A.C., Reiner, A.P., Ridker, P.M., Rotter, J.I., Toniolo, D.,
Uitterlinden, A.G., Ulivi, S., Völzke, H., Wareham, N.J., Weir, D.R., Yerges-
Armstrong, L.M., PRACTICAL consortium, kConFab Investigators, AOCS
Investigators, Generation Scotland, EPIC-InterAct Consortium, LifeLines Cohort
Study, Price, A.L., Stefansson, K., Visser, J.A., Ong, K.K., Chang-Claude, J., Murabito,
J.M., Perry, J.R.B., Murray, A., 2015. Large-scale genomic analyses link reproductive
aging to hypothalamic signaling, breast cancer susceptibility and BRCA1-mediated
DNA repair. Nat. Genet. 47, 12941303.
de Almeida, A.J.P.O., Ribeiro, T.P., de Medeiros, I.A., 2017. Aging: Molecular Pathways
and Implications on the Cardiovascular System. Oxid. Med. Cell. Longev. https://doi.
org/10.1155/2017/7941563. 2017.
de Bock, M., Derraik, J.G.B., Brennan, C.M., Biggs, J.B., Morgan, P.E., Hodgkinson, S.C.,
Hofman, P.L., Cuteld, W.S., 2013. Olive (Olea europaea L.) leaf polyphenols im-
prove insulin sensitivity in middle-aged overweight men: a randomized, placebo-
controlled, crossover trial. PLoS One 8, e57622.
de Courten, B., Jakubova, M., de Courten, M.P., Kukurova, I.J., Vallova, S., Krumpolec, P.,
Valkovic, L., Kurdiova, T., Garzon, D., Barbaresi, S., Teede, H.J., Derave, W., Krssak,
M., Aldini, G., Ukropec, J., Ukropcova, B., 2016. Eects of carnosine supplementation
on glucose metabolism: Pilot clinical trial. Obesity 24, 10271034.
Deelen, J., Evans, D.S., Arking, D.E., Tesi, N., Nygaard, M., Liu, X., Wojczynski, M.K.,
Biggs, M.L., van der Spek, A., Atzmon, G., Ware, E.B., Sarnowski, C., Smith, A.V.,
Seppälä, I., Cordell, H.J., Dose, J., Amin, N., Arnold, A.M., Ayers, K.L., Barzilai, N.,
Becker, E.J., Beekman, M., Blanché, H., Christensen, K., Christiansen, L., Collerton,
J.C., Cubaynes, S., Cummings, S.R., Davies, K., Debrabant, B., Deleuze, J.-F., Duncan,
R., Faul, J.D., Franceschi, C., Galan, P., Gudnason, V., Harris, T.B., Huisman, M.,
Hurme, M.A., Jagger, C., Jansen, I., Jylhä, M., Kähönen, M., Karasik, D., Kardia,
S.L.R., Kingston, A., Kirkwood, T.B.L., Launer, L.J., Lehtimäki, T., Lieb, W.,
Lyytikäinen, L.-P., Martin-Ruiz, C., Min, J., Nebel, A., Newman, A.B., Nie, C., Nohr,
E.A., Orwoll, E.S., Perls, T.T., Province, M.A., Psaty, B.M., Raitakari, O.T., Reinders,
M.J.T., Robine, J.-M., Rotter, J.I., Sebastiani, P., Smith, J., Sørensen, T.I.A., Taylor,
K.D., Uitterlinden, A.G., van der Flier, W., van der Lee, S.J., van Duijn, C.M., van
Heemst, D., Vaupel, J.W., Weir, D., Ye, K., Zeng, Y., Zheng, W., Holstege, H., Kiel,
D.P., Lunetta, K.L., Slagboom, P.E., Murabito, J.M., 2019. A meta-analysis of genome-
wide association studies identies multiple longevity genes. Nat. Commun. 10, 3669.
de Grey, A., Rae, M., 2007. Ending aging: The rejuvenation breakthroughs that could
reverse human aging in our lifetime. St. Martins Press, NY.
de Kreutzenberg, S.V., Ceolotto, G., Cattelan, A., Pagnin, E., Mazzucato, M., Garagnani,
P., Borelli, V., Bacalini, M.G., Franceschi, C., Fadini, G.P., Avogaro, A., 2015.
Metformin improves putative longevity eectors in peripheral mononuclear cells
from subjects with prediabetes. A randomized controlled trial. Nutr. Metab.
Cardiovasc. Dis. 25, 686693.
Delgado, T.C., 2013. Glutamate and GABA in Appetite Regulation. Front. Endocrinol. 4,
103.
de Magalhães, J.P., Curado, J., Church, G.M., 2009. Meta-analysis of age-related gene
expression proles identies common signatures of aging. Bioinformatics 25,
875881.
Despa, S., Margulies, K.B., Chen, L., Knowlton, A.A., Havel, P.J., Taegtmeyer, H., Bers,
D.M., Despa, F., 2012. Hyperamylinemia contributes to cardiac dysfunction in obesity
and diabetes: a study in humans and rats. Circ. Res. 110, 598608.
Deutz, N.E., Matheson, E.M., Matarese, L.E., Luo, M., Baggs, G.E., Nelson, J.L., Hegazi,
R.A., Tappenden, K.A., Ziegler, T.R., NOURISH Study Group, 2016. Readmission and
mortality in malnourished, older, hospitalized adults treated with a specialized oral
nutritional supplement: A randomized clinical trial. Clin. Nutr. 35, 1826.
De Vecchis, R., Cesaro, A., Ariano, C., Giasi, A., Cioppa, C., 2017. Phosphodiesterase-5
Inhibitors Improve Clinical Outcomes, Exercise Capacity and Pulmonary
Hemodynamics in Patients With Heart Failure With Reduced Left Ventricular Ejection
Fraction: A Meta-Analysis. J. Clin. Med. Res. 9, 488.
Dewundara, S.S., Wiggs, J.L., Sullivan, D.A., Pasquale, L.R., 2016. Is Estrogen a
Therapeutic Target for Glaucoma? Semin. Ophthalmol. 31, 140146.
COMBO ENDO TEAM: 2016,Diamanti-Kandarakis, E., Dattilo, M., Macut, D., Duntas, L.,
Gonos, E.S., Goulis, D.G., Gantenbein, C.K., Kapetanou, M., Koukkou, E.,
Lambrinoudaki, I., Michalaki, M., Eftekhari-Nader, S., Pasquali, R., Peppa, M.,
Tzanela, M., Vassilatou, E., Vryonidou, A., 2017. Mechanisms in endocrionology:
aging and anti-aging: a combo-endocrinology overview. Eur. J. Endocrinol. 176 (6),
R283R308. https://doi.org/10.1530/EJE-16-1061.
Ding, N., Nie, H., Sun, X., Sun, W., Qu, Y., Liu, X., Yao, Y., Liang, X., Chen, C.C., Li, Y.,
2011. Human serum N-glycan proles are age and sex dependent. Age Ageing 40,
568575.
Di Paola, M., Lorusso, M., 2006. Interaction of free fatty acids with mitochondria: cou-
pling, uncoupling and permeability transition. Biochim. Biophys. Acta 1757,
13301337.
Domecq, J.P., Prutsky, G., Leppin, A., Sonbol, M.B., Altayar, O., Undavalli, C., Wang, Z.,
Elraiyah, T., Brito, J.P., Mauck, K.F., Lababidi, M.H., Prokop, L.J., Asi, N., Wei, J.,
Fidahussein, S., Montori, V.M., Murad, M.H., 2015. Clinical review: Drugs commonly
associated with weight change: a systematic review and meta-analysis. J. Clin.
Endocrinol. Metab. 100, 363370.
Doosti-Irani, A., Ostadmohammadi, V., Mirhosseini, N., Mansournia, M., Reiter, R.J.,
Kashanian, M., Rahimi, M., Razavi, M., Asemi, Z., 2018. The Eects of Melatonin
Supplementation on Glycemic Control: A Systematic Review and Meta-Analysis of
Randomized Controlled Trials. Hormone and Metabolic Research 50, 783790.
Dorfman, M.D., Thaler, J.P., 2015. Hypothalamic inammation and gliosis in obesity.
Curr. Opin. Endocrinol. Diabetes Obes. 22, 325330.
Doria, E., Buonocore, D., Focarelli, A., Marzatico, F., 2012. Relationship between human
aging muscle and oxidative system pathway. Oxid. Med. Cell. Longev., 830257 2012.
Dow, C.T., Harley, C.B., 2016. Evaluation of an oral telomerase activator for early age-
related macular degeneration - a pilot study. Clin. Ophthalmol. 10, 243249.
Draper, J., Webb, J., Jackson, T., Jones, H., Rinaldi, C.A., Schi, R., McDonagh, T.,
Razavi, R., S Carr-White, G., 2018. Comparison of the Diagnostic Accuracy of Plasma
N-Terminal Pro-Brain Natriuretic Peptide in Patients 80 Years of Age with Heart
Failure. Am. J. Cardiol. 122, 20752079.
Drougard, A., Fournel, A., Valet, P., Knauf, C., 2015. Impact of hypothalamic reactive
oxygen species in the regulation of energy metabolism and food intake. Front.
Neurosci. 9, 56.
Duca, L., Blaise, S., Romier, B., Laargue, M., Gayral, S., El Btaouri, H., Kawecki, C.,
Guillot, A., Martiny, L., Debelle, L., Maurice, P., 2016. Matrix ageing and vascular
impacts: focus on elastin fragmentation. Cardiovasc. Res. 110, 298308.
Dudinskaya, E.N., Tkacheva, O.N., Shestakova, M.V., Brailova, N.V., Strazhesko, I.D.,
Akasheva, D.U., Isaykina, O.Y., Sharashkina, N.V., Kashtanova, D.A., Boytsov, S.A.,
2015. Short telomere length is associated with arterial aging in patients with type 2
diabetes mellitus. Endocr Connect 4, 136143.
Duggal, N.A., Pollock, R.D., Lazarus, N.R., Harridge, S., Lord, J.M., 2018. Major features
of immunesenescence, including reduced thymic output, are ameliorated by high
levels of physical activity in adulthood. Aging Cell 17. https://doi.org/10.1111/acel.
12750.
El-Sakka, A.I., 2011. Reversion of penile brosis: Current information and a new horizon.
Arab Journal of Urology 9, 4955.
Emanuele, E., Minoretti, P., Sanchis-Gomar, F., Pareja-Galeano, H., Yilmaz, Y.,
Garatachea, N., Lucia, A., 2014. Can enhanced autophagy be associated with human
longevity? Serum levels of the autophagy biomarker beclin-1 are increased in healthy
centenarians. Rejuvenation Res. 17, 518524.
Ettehad, D., Emdin, C.A., Kiran, A., Anderson, S.G., Callender, T., Emberson, J., Chalmers,
J., Rodgers, A., Rahimi, K., 2016. Blood pressure lowering for prevention of cardio-
vascular disease and death: a systematic review and meta-analysis. Lancet 387,
957967.
Evans, C.J., Ho, Y., Daveson, B.A., Hall, S., Higginson, I.J., Gao, W., GUIDE_Care project,
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
18
2014. Place and cause of death in centenarians: a population-based observational
study in England, 2001 to 2010. PLoS Med. 11, e1001653.
Fabiani, R., 2016. Anti-cancer properties of olive oil secoiridoid phenols: a systematic
review of in vivo studies. Food Funct. 7, 41454159.
Fahy, G.M., Brooke, R.T., Watson, J.P., Good, Z., Vasanawala, S.S., Maecker, H., Leipold,
M.D., Lin, D.T.S., Kobor, M.S., Horvath, S., 2019. Reversal of epigenetic aging and
immunosenescent trends in humans. Aging Cell, e13028.
Fang, X., Liang, C., Li, M., Montgomery, S., Fall, K., Aaseth, J., Cao, Y., 2016. Dose-
response relationship between dietary magnesium intake and cardiovascular mor-
tality: A systematic review and dose-based meta-regression analysis of prospective
studies. J. Trace Elem. Med. Biol. 38, 6473.
Farrokhian, A., Raygan, F., Bahmani, F., Talari, H.R., Esfandiari, R., Esmaillzadeh, A.,
Asemi, Z., 2017. Long-Term Vitamin D Supplementation Aects Metabolic Status in
Vitamin D-Decient Type 2 Diabetic Patients with Coronary Artery Disease. J. Nutr.
147, 384389.
Fatima, S., Jameel, A., Ayesha, F., Menzies, D.J., 2017. The shifting paradigm in the
treatment of type 2 diabetes mellitus-A cardiologists perspective. Clin. Cardiol. 40,
970973.
Fedewa, M.V., Hathaway, E.D., Ward-Ritacco, C.L., 2017. Eect of exercise training on C
reactive protein: a systematic review and meta-analysis of randomised and non-
randomised controlled trials. Br. J. Sports Med. 51, 670676.
Ferrario, C.R., Labouèbe, G., Liu, S., Nieh, E.H., Routh, V.H., Xu, S., OConnor, E.C., 2016.
Homeostasis Meets Motivation in the Battle to Control Food Intake. J. Neurosci. 36,
1146911481.
Filip, R., Possemiers, S., Heyerick, A., Pinheiro, I., Raszewski, G., Davicco, M.-J., Coxam,
V., 2015. Twelve-month consumption of a polyphenol extract from olive (Olea
europaea) in a double blind, randomized trial increases serum total osteocalcin levels
and improves serum lipid proles in postmenopausal women with osteopenia. J.
Nutr. Health Aging 19, 7786.
Fiorito, G., McCrory, C., Robinson, O., Carmeli, C., Rosales, C.O., Zhang, Y., Colicino, E.,
Dugué, P.-A., Artaud, F., McKay, G.J., Jeong, A., Mishra, P.P., Nøst, T.H., Krogh, V.,
Panico, S., Sacerdote, C., Tumino, R., Palli, D., Matullo, G., Guarrera, S., Gandini, M.,
Bochud, M., Dermitzakis, E., Muka, T., Schwartz, J., Vokonas, P.S., Just, A., Hodge,
A.M., Giles, G.G., Southey, M.C., Hurme, M.A., Young, I., McKnight, A.J., Kunze, S.,
Waldenberger, M., Peters, A., Schwettmann, L., Lund, E., Baccarelli, A., Milne, R.L.,
Kenny, R.A., Elbaz, A., Brenner, H., Kee, F., Voortman, T., Probst-Hensch, N.,
Lehtimäki, T., Elliot, P., Stringhini, S., Vineis, P., Polidoro, S., BIOS Consortium,
Lifepath consortium, 2019. Socioeconomic position, lifestyle habits and biomarkers
of epigenetic aging: a multi-cohort analysis. Aging 11, 20452070.
Firuzi, O., Miri, R., Tavakkoli, M., Saso, L., 2011. Antioxidant therapy: current status and
future prospects. Curr. Med. Chem. 18, 38713888.
Forero, D.A., González-Giraldo, Y., López-Quintero, C., Castro-Vega, L.J., Barreto, G.E.,
Perry, G., 2016. Meta-analysis of Telomere Length in Alzheimers Disease. J.
Gerontol. A Biol. Sci. Med. Sci. 71, 10691073.
Forman, D.E., Berman, A.D., McCabe, C.H., Baim, D.S., Wei, J.Y., 1992. PTCA in the
elderly: the young-oldversus the old-old.. J. Am. Geriatr. Soc. 40, 1922.
Franceschi, C., Campisi, J., 2014. Chronic inammation (inammaging) and its potential
contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 69 (Suppl
1), S49.
Francis-Emmanuel, P.M., Thompson, D.S., Barnett, A.T., Osmond, C., Byrne, C.D.,
Hanson, M.A., Gluckman, P.D., Forrester, T.E., Boyne, M.S., 2014. Glucose metabo-
lism in adult survivors of severe acute malnutrition. J. Clin. Endocrinol. Metab. 99,
22332240.
Franco, I., Johansson, A., Olsson, K., Vrtačnik, P., Lundin, P., Helgadottir, H.T., Larsson,
M., Revêchon, G., Bosia, C., Pagnani, A., Provero, P., Gustafsson, T., Fischer, H.,
Eriksson, M., 2018. Somatic mutagenesis in satellite cells associates with human
skeletal muscle aging. Nat. Commun. 9. https://doi.org/10.1038/s41467-018-
03244-6.
Fransquet, P.D., Wrigglesworth, J., Woods, R.L., Ernst, M.E., Ryan, J., 2019. The epige-
netic clock as a predictor of disease and mortality risk: a systematic review and meta-
analysis. Clin. Epigenetics 11, 62.
Freund, A., Orjalo, A.V., Desprez, P.-Y., Campisi, J., 2010. Inammatory networks during
cellular senescence: causes and consequences. Trends Mol. Med. 16, 238246.
Fried, L.P., Tangen, C.M., Walston, J., Newman, A.B., Hirsch, C., Gottdiener, J., Seeman,
T., Tracy, R., Kop, W.J., Burke, G., McBurnie, M.A., Cardiovascular Health Study
Collaborative Research Group, 2001. Frailty in older adults: evidence for a pheno-
type. J. Gerontol. A Biol. Sci. Med. Sci. 56, M14656.
Fukushima, Y., Daida, H., Morimoto, T., Kasai, T., Miyauchi, K., Yamagishi, S.-I.,
Takeuchi, M., Hiro, T., Kimura, T., Nakagawa, Y., Yamagishi, M., Ozaki, Y.,
Matsuzaki, M., JAPAN-ACS Investigators, 2013. Relationship between advanced
glycation end products and plaque progression in patients with acute coronary syn-
drome: the JAPAN-ACS sub-study. Cardiovasc. Diabetol. 12, 5.
Gaddipati, R., Sasikala, M., Padaki, N., Mukherjee, R.M., Sekaran, A., Jayaraj-Mansard,
M., Rabella, P., Rao-Guduru, V., Reddy-Duvvuru, N., 2010. Visceral adipose tissue
visfatin in nonalcoholic fatty liver disease. Ann. Hepatol. 9, 266270.
Gangisetty, O., Cabrera, M.A., Murugan, S., 2018. Impact of epigenetics in aging and age
related neurodegenerative diseases. Front. Biosci. 23, 14451464.
Gao, X., Colicino, E., Shen, J., Just, A.C., Nwanaji-Enwerem, J.C., Wang, C., Coull, B., Lin,
X., Vokonas, P., Zheng, Y., Hou, L., Schwartz, J., Baccarelli, A.A., 2019. Comparative
validation of an epigenetic mortality risk score with three aging biomarkers for
predicting mortality risks among older adult males. Int. J. Epidemiol. 48, 19581971.
Garg, K., Corona, B.T., Walters, T.J., 2015. Therapeutic strategies for preventing skeletal
muscle brosis after injury. Front. Pharmacol. 6, 87.
Gauthier, A.C., Liu, J., 2017. Epigenetics and Signaling Pathways in Glaucoma. Biomed
Res. Int., 5712341 2017.
GBD 2017 Risk Factor Collaborators, 2018. Global, regional, and national comparative
risk assessment of 84 behavioural, environmental and occupational, and metabolic
risks or clusters of risks for 195 countries and territories, 1990-2017: a systematic
analysis for the Global Burden of Disease Study 2017. Lancet 392, 19231994.
Geifman, N., Brinton, R.D., Kennedy, R.E., Schneider, L.S., Butte, A.J., 2017. Evidence for
benet of statins to modify cognitive decline and risk in Alzheimers disease.
Alzheimers. Res. Ther. 9, 10.
Giannandrea, M., Parks, W.C., 2014. Diverse functions of matrix metalloproteinases
during brosis. Dis. Model. Mech. 7, 193203.
Giannoulis, M.G., Martin, F.C., Sreekumaran Nair, K., Margot Umpleby, A., Sonksen, P.,
2012. Hormone Replacement Therapy and Physical Function in Healthy Older Men.
Time to Talk Hormones? Endocr. Rev. 33, 314377.
Giezenaar, C., Chapman, I., Luscombe-Marsh, N., Feinle-Bisset, C., Horowitz, M., Soenen,
S., 2016. Ageing Is Associated with Decreases in Appetite and Energy IntakeA
Meta-Analysis in Healthy Adults. Nutrients 8, 28.
Giorgi, C., Bonora, M., Sorrentino, G., Missiroli, S., Poletti, F., Suski, J.M., Galindo
Ramirez, F., Rizzuto, R., Di Virgilio, F., Zito, E., Pandol, P.P., Wieckowski, M.R.,
Mammano, F., Del Sal, G., Pinton, P., 2015. p53 at the endoplasmic reticulum reg-
ulates apoptosis in a Ca2+-dependent manner. Proc. Natl. Acad. Sci. U. S. A. 112,
17791784.
Giuseppe, C., Ross, J.M., 2016. Mitochondria in Ageing and Diseases: The Super Trouper
of the Cell. Int. J. Mol. Sci. 17. https://doi.org/10.3390/ijms17050711.
Global BMI Mortality Collaboration, Di Angelantonio, E., Bhupathiraju, S., Wormser, D.,
Gao, P., Kaptoge, S., Berrington de Gonzalez, A., Cairns, B., Huxley, R., Jackson, C.,
Joshy, G., Lewington, S., Manson, J., Murphy, N., Patel, A., Samet, J., Woodward, M.,
Zheng, W., Zhou, M., Bansal, N., Barricarte, A., Carter, B., Cerhan, J., Smith, G., Fang,
X., Franco, O., Green, J., Halsey, J., Hildebrand, J., Jung, K., Korda, R., McLerran, D.,
Moore, S., OKeee, L., Paige, E., Ramond, A., Reeves, G., Rolland, B., Sacerdote, C.,
Sattar, N., Soanopoulou, E., Stevens, J., Thun, M., Ueshima, H., Yang, L., Yun, Y.,
Willeit, P., Banks, E., Beral, V., Chen, Z., Gapstur, S., Gunter, M., Hartge, P., Jee, S.,
Lam, T.-H., Peto, R., Potter, J., Willett, W., Thompson, S., Danesh, J., Hu, F., 2016.
Body-mass index and all-cause mortality: individual-participant-data meta-analysis of
239 prospective studies in four continents. Lancet 388, 776786.
Golzarand, M., Shab-Bidar, S., Koochakpoor, G., Speakman, J.R., Djafarian, K., 2016.
Eect of vitamin D3 supplementation on blood pressure in adults: An updated meta-
analysis. Nutr. Metab. Cardiovasc. Dis. 26, 663673.
Gong, J., Fang, K., Dong, H., Wang, D., Hu, M., Lu, F., 2016. Eect of fenugreek on
hyperglycaemia and hyperlipidemia in diabetes and prediabetes: A meta-analysis. J.
Ethnopharmacol. 194, 260268.
Gong, J., Qin, X., Yuan, F., Hu, M., Chen, G., Fang, K., Wang, D., Jiang, S., Li, J., Zhao, Y.,
Huang, Z., Dong, H., Lu, F., 2018. Ecacy and safety of sugarcane policosanol on
dyslipidemia: A meta-analysis of randomized controlled trials. Mol. Nutr. Food Res.
62. https://doi.org/10.1002/mnfr.201700280.
Gopalan, S., Carja, O., Fagny, M., Patin, E., Myrick, J.W., McEwen, L.M., Mah, S.M.,
Kobor, M.S., Froment, A., Feldman, M.W., Quintana-Murci, L., Henn, B.M., 2017.
Trends in DNA Methylation with Age Replicate Across Diverse Human Populations.
Genetics 206, 16591674.
Gracia, K.C., Llanas-Cornejo, D., Husi, H., 2017. CVD and Oxidative Stress. J. Clin. Med.
Res. 6. https://doi.org/10.3390/jcm6020022.
Grady, D.L., Thanos, P.K., Corrada, M.M., Barnett Jr, J.C., Ciobanu, V., Shustarovich, D.,
Napoli, A., Moyzis, A.G., Grandy, D., Rubinstein, M., Wang, G.-J., Kawas, C.H., Chen,
C., Dong, Q., Wang, E., Volkow, N.D., Moyzis, R.K., 2013. DRD4 genotype predicts
longevity in mouse and human. J. Neurosci. 33, 286291.
Grant, W.B., 2018. A Review of the Evidence Supporting the Vitamin D-Cancer Prevention
Hypothesis in 2017. Anticancer Res. 38, 11211136.
Gretchen Garbe Collins, B.V.R., 2015. The impact of lifestyle modications, diet, and
vitamin supplementation on natural fertility. Fertility Research and Practice 1.
https://doi.org/10.1186/s40738-015-0003-4.
Guest, J., Grant, R., Mori, T.A., Croft, K.D., 2014. Changes in oxidative damage, in-
ammation and [NAD(H)] with age in cerebrospinal uid. PLoS One 9, e85335.
Gunes, S., Hekim, G.N.T., Arslan, M.A., Asci, R., 2016. Eects of aging on the male re-
productive system. J. Assist. Reprod. Genet. 33, 441454.
Guo, L., Zhang, S., 2017. Association between ideal cardiovascular health metrics and risk
of cardiovascular events or mortality: A meta-analysis of prospective studies. Clin.
Cardiol. 40, 13391346.
Guo, X.-F., Li, J.-M., Tang, J., Li, D., 2017. Eects of resveratrol supplementation on risk
factors of non-communicable diseases: A meta-analysis of randomized controlled
trials. Crit. Rev. Food Sci. Nutr. 115.
Hackett, G., 2016. An update on the role of testosterone replacement therapy in the
management of hypogonadism. Ther. Adv. Urol. 8, 147160.
Hackett, G., Jones, P.W., Strange, R.C., Ramachandran, S., 2017. Statin, testosterone and
phosphodiesterase 5-inhibitor treatments and age related mortality in diabetes.
World J. Diabetes 8, 104.
Hannum, G., Guinney, J., Zhao, L., Zhang, L., Hughes, G., Sadda, S., Klotzle, B., Bibikova,
M., Fan, J.-B., Gao, Y., Deconde, R., Chen, M., Rajapakse, I., Friend, S., Ideker, T.,
Zhang, K., 2013. Genome-wide methylation proles reveal quantitative views of
human aging rates. Mol. Cell 49, 359367.
Hanson, A.J., Bayer-Carter, J.L., Green, P.S., Montine, T.J., Wilkinson, C.W., Baker, L.D.,
Watson, G.S., Bonner, L.M., Callaghan, M., Leverenz, J.B., Tsai, E., Postupna, N.,
Zhang, J., Lampe, J., Craft, S., 2013. Eect of apolipoprotein E genotype and diet on
apolipoprotein E lipidation and amyloid peptides: randomized clinical trial. JAMA
Neurol. 70, 972980.
Hardeland, R., Cardinali, D.P., Brown, G.M., Pandi-Perumal, S.R., 2015. Melatonin and
brain inammaging. Prog. Neurobiol. 127-128, 4663.
Harlow, S.D., Gass, M., Hall, J.E., Lobo, R., Maki, P., Rebar, R.W., Sherman, S., Sluss,
P.M., de Villiers, T.J., STRAW + 10 Collaborative Group, 2012. Executive summary
of the Stages of Reproductive Aging Workshop + 10: addressing the unnished
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
19
agenda of staging reproductive aging. Fertil. Steril. 97, 843851.
Haukka, J., Niskanen, L., Auvinen, A., 2017. Risk of Cause-Specic Death in Individuals
with Cancer-Modifying Role Diabetes, Statins and Metformin. Int. J. Cancer 141,
24372449.
Haycock, P.C., Heydon, E.E., Kaptoge, S., Butterworth, A.S., Thompson, A., Willeit, P.,
2014. Leucocyte telomere length and risk of cardiovascular disease: systematic re-
view and meta-analysis. BMJ 349, g4227.
He, H., Zhou, X., Wang, Q., Zhao, Y., 2013. Does the Couse of Astragalus-Containing
Chinese Herbal Prescriptions and Radiotherapy Benet to Non-Small-Cell Lung
Cancer Treatment: A Meta-Analysis of Randomized Trials. Evid. Based. Complement.
Alternat. Med. https://doi.org/10.1155/2013/426207. 2013.
Heinonen, S., Saarinen, L., Naukkarinen, J., Rodríguez, A., Frühbeck, G., Hakkarainen, A.,
Lundbom, J., Lundbom, N., Vuolteenaho, K., Moilanen, E., Arner, P., Hautaniemi, S.,
Suomalainen, A., Kaprio, J., Rissanen, A., Pietiläinen, K.H., 2014. Adipocyte mor-
phology and implications for metabolic derangements in acquired obesity. Int. J.
Obes. 38, 14231431.
He, S., Sharpless, N.E., 2017. Senescence in Health and Disease. Cell 169, 10001011.
Hisatsune, T., Kaneko, J., Kurashige, H., Cao, Y., Satsu, H., Totsuka, M., Katakura, Y.,
Imabayashi, E., Matsuda, H., 2016. Eect of Anserine/Carnosine Supplementation on
Verbal Episodic Memory in Elderly People. J. Alzheimers. Dis. 50, 149159.
Ho, A., Gabriel, A., Bhatnagar, A., Etienne, D., Loukas, M., 2014. Seasonality pattern of
breast, colorectal, and prostate cancer is dependent on latitude. Med. Sci. Monit. 20,
818824.
Hochschild, R., 1989. Improving the precision of biological age determinations. Part 1: A
new approach to calculating biological age. Exp. Gerontol. 24, 289300.
Hodes, R.J., Sierra, F., Austad, S.N., Epel, E., Neigh, G.N., Erlandson, K.M., Schafer, M.J.,
LeBrasseur, N.K., Wiley, C., Campisi, J., Sehl, M.E., Scalia, R., Eguchi, S., Kasinath,
B.S., Halter, J.B., Cohen, H.J., Demark-Wahnefried, W., Ahles, T.A., Barzilai, N.,
Hurria, A., Hunt, P.W., 2016. Disease drivers of aging. Ann. N. Y. Acad. Sci. 1386, 45.
Hoel, D.G., Berwick, M., de Gruijl, F.R., Holick, M.F., 2016. The risks and benets of sun
exposure 2016. Dermatoendocrinol 8. https://doi.org/10.1080/19381980.2016.
1248325.
Hostedt, J., Arner, E., Wahrenberg, H., Andersson, D.P., Qvisth, V., Löfgren, P., Rydén,
M., Thörne, A., Wirén, M., Palmér, M., Thorell, A., Toft, E., Arner, P., 2010. Regional
impact of adipose tissue morphology on the metabolic prole in morbid obesity.
Diabetologia 53, 24962503.
Holly, A.C., Melzer, D., Pilling, L.C., Henley, W., Hernandez, D.G., Singleton, A.B.,
Bandinelli, S., Guralnik, J.M., Ferrucci, L., Harries, L.W., 2013. Towards a gene ex-
pression biomarker set for human biological age. Aging Cell 12, 324326.
Honda, H., Hosaka, N., Aoshima, Y., Hirai, Y., Michihata, T., Akizawa, T., 2012.
Olmesartan medoxomil is associated with decreased plasma AGEs, pentosidine, and
N-(epsilon)-carboxymethyl-lysine levels in hemodialysis patients. Clin. Exp.
Hypertens. 34, 1723.
Horvath, S., 2013. DNA methylation age of human tissues and cell types. Genome Biol.
14, R115.
Horvath, S., Oshima, J., Martin, G.M., Lu, A.T., Quach, A., Cohen, H., Felton, S.,
Matsuyama, M., Lowe, D., Kabacik, S., Wilson, J.G., Reiner, A.P., Maierhofer, A.,
Flunkert, J., Aviv, A., Hou, L., Baccarelli, A.A., Li, Y., Stewart, J.D., Whitsel, E.A.,
Ferrucci, L., Matsuyama, S., Raj, K., 2018. Epigenetic clock for skin and blood cells
applied to Hutchinson Gilford Progeria Syndrome and studies. Aging 10, 17581775.
Hotamisligil, G.S., Arner, P., Atkinson, R.L., Spiegelman, B.M., 1997. Dierential reg-
ulation of the p80 tumor necrosis factor receptor in human obesity and insulin re-
sistance. Diabetes 46, 451455.
Houmard, J.A., Tanner, C.J., Yu, C., Cunningham, P.G., Pories, W.J., MacDonald, K.G.,
Shulman, G.I., 2002. Eect of weight loss on insulin sensitivity and intramuscular
long-chain fatty acyl-CoAs in morbidly obese subjects. Diabetes 51, 29592963.
Huang, R.-Y., Huang, C.-C., Hu, F.B., Chavarro, J.E., 2016a. Vegetarian Diets and Weight
Reduction: a Meta-Analysis of Randomized Controlled Trials. J. Gen. Intern. Med. 31,
109116.
Huang, Y., Cai, X., Mai, W., Li, M., Hu, Y., 2016b. Association between prediabetes and
risk of cardiovascular disease and all cause mortality: systematic review and meta-
analysis. BMJ 355, i5953.
Hu, H.-H., Chen, D.-Q., Wang, Y.-N., Feng, Y.-L., Cao, G., Vaziri, N.D., Zhao, Y.-Y., 2018.
New insights into TGF-β/Smad signaling in tissue brosis. Chem. Biol. Interact. 292,
7683.
Hunter, J.M., Kwan, J., Malek-Ahmadi, M., Maarouf, C.L., Kokjohn, T.A., Belden, C.,
Sabbagh, M.N., Beach, T.G., Roher, A.E., 2012. Morphological and Pathological
Evolution of the Brain Microcirculation in Aging and Alzheimers Disease. PLoS One
7. https://doi.org/10.1371/journal.pone.0036893.
Iannuzzi, C., Irace, G., Sirangelo, I., 2014. Dierential eects of glycation on protein
aggregation and amyloid formation. Frontiers in Molecular Biosciences 1. https://
doi.org/10.3389/fmolb.2014.00009.
Ibáñez-Sanz, G., Díez-Villanueva, A., Vilorio-Marqués, L., Gracia, E., Aragonés, N.,
Olmedo-Requena, R., Llorca, J., Vidán, J., Amiano, P., Nos, P., Fernández-Tardón, G.,
Rada, R., Chirlaque, M.D., Guinó, E., Dávila-Batista, V., Castaño-Vinyals, G., Pérez-
Gómez, B., Mirón-Pozo, B., Dierssen-Sotos, T., Etxeberria, J., Molinuevo, A., Álvarez-
Cuenllas, B., Kogevinas, M., Pollán, M., Moreno, V., 2018. Possible role of chondroitin
sulphate and glucosamine for primary prevention of colorectal cancer. Results from
the MCC-Spain study. Sci. Rep. 8, 2040.
Irvin, M.R., Aslibekyan, S., Do, A., Zhi, D., Hidalgo, B., Claas, S.A., Srinivasasainagendra,
V., Horvath, S., Tiwari, H.K., Absher, D.M., Arnett, D.K., 2018. Metabolic and in-
ammatory biomarkers are associated with epigenetic aging acceleration estimates in
the GOLDN study. Clin. Epigenetics 10, 56.
Ishikawa, N., Nakamura, K.-I., Izumiyama-Shimomura, N., Aida, J., Matsuda, Y., Arai, T.,
Takubo, K., 2016. Changes of telomere status with aging: An update. Geriatr.
Gerontol. Int. 16 (Suppl 1), 3042.
Ismail, M.N., Chee, S.S., Nawawi, H., Yuso, K., Lim, T.O., James, W.P.T., 2002. Obesity
in Malaysia. Obesity Reviews. https://doi.org/10.1046/j.1467-789x.2002.00074.x.
Jafari, T., Fallah, A.A., Barani, A., 2016. Eects of vitamin D on serum lipid prole in
patients with type 2 diabetes: A meta-analysis of randomized controlled trials. Clin.
Nutr. 35, 12591268.
Jaganathan, R., Ravindran, R., Dhanasekaran, S., 2018. Emerging Role of Adipocytokines
in Type 2 Diabetes as Mediators of Insulin Resistance and Cardiovascular Disease.
Can J Diabetes 42, 446456 e1.
Jaiswal, S., Natarajan, P., Silver, A.J., Gibson, C.J., Bick, A.G., Shvartz, E., McConkey, M.,
Gupta, N., Gabriel, S., Ardissino, D., Baber, U., Mehran, R., Fuster, V., Danesh, J.,
Frossard, P., Saleheen, D., Melander, O., Sukhova, G.K., Neuberg, D., Libby, P.,
Kathiresan, S., Ebert, B.L., 2017. Clonal Hematopoiesis and Risk of Atherosclerotic
Cardiovascular Disease. N. Engl. J. Med. 377, 111121.
Jeong, H.S., Lee, D.W., Park, K.H., Lee, Y.K., Bae, S.H., Kang, M.J., Shim, M.S., Teong,
C.H., 2013. Clinical Factors Related to Frailty Estimated by the Korean Frailty Index.
Journal of the Korean Geriatrics Society 17, 71.
Jeykodi, S., Deshpande, J., Juturu, V., 2016. Salacia Extract Improves Postprandial
Glucose and Insulin Response: A Randomized Double-Blind, Placebo Controlled,
Crossover Study in Healthy Volunteers. J Diabetes Res, 7971831 2016.
Jia, L., Zhang, W., Chen, X., 2017. Common methods of biological age estimation. Clin.
Interv. Aging 12, 759.
Jin, S., Diano, S., 2018. Mitochondrial Dynamics and Hypothalamic Regulation of
Metabolism. Endocrinology 159, 35963604.
Johnstone, J., Parsons, R., Botelho, F., Millar, J., McNeil, S., Fulop, T., McElhaney, J.E.,
Andrew, M.K., Walter, S.D., Devereaux, P.J., Malek, M., Brinkman, R.R., Bramson, J.,
Loeb, M., 2017. T-Cell Phenotypes Predictive of Frailty and Mortality in Elderly
Nursing Home Residents. J. Am. Geriatr. Soc. 65, 153159.
Jones, C.M., Boelaert, K., 2015. The Endocrinology of Ageing: A Mini-Review.
Gerontology 61, 291300.
Jung, S.-E., Lim, S.M., Hong, S.R., Lee, E.H., Shin, K.-J., Lee, H.Y., 2019. DNA methylation
of the ELOVL2, FHL2, KLF14, C1orf132/MIR29B2C, and TRIM59 genes for age
prediction from blood, saliva, and buccal swab samples. Forensic Sci. Int. Genet.
38, 18.
Jura, M., Kozak, L.P., 2016. Obesity and related consequences to ageing. Age 38. https://
doi.org/10.1007/s11357-016-9884-3.
Justice, J.N., Nambiar, A.M., Tchkonia, T., LeBrasseur, N.K., Pascual, R., Hashmi, S.K.,
Prata, L., Masternak, M.M., Kritchevsky, S.B., Musi, N., Kirkland, J.L., 2019.
Senolytics in idiopathic pulmonary brosis: Results from a rst-in-human, open-label,
pilot study. EBioMedicine. https://doi.org/10.1016/j.ebiom.2018.12.052.
Kahal, H., Abouda, G., Rigby, A.S., Coady, A.M., Kilpatrick, E.S., Atkin, S.L., 2014.
Glucagon-like peptide-1 analogue, liraglutide, improves liver brosis markers in
obese women with polycystic ovary syndrome and nonalcoholic fatty liver disease.
Clin. Endocrinol. 81, 523528.
Kang, H.M., Ahn, S.H., Choi, P., Ko, Y.-A., Han, S.H., Chinga, F., Park, A.S.D., Tao, J.,
Sharma, K., Pullman, J., Bottinger, E.P., Goldberg, I.J., Susztak, K., 2014. Defective
fatty acid oxidation in renal tubular epithelial cells has a key role in kidney brosis
development. Nat. Med. 21, 3746.
Kantor, E.D., Zhang, X., Wu, K., Signorello, L.B., Chan, A.T., Fuchs, C.S., Giovannucci,
E.L., 2016. Use of glucosamine and chondroitin supplements in relation to risk of
colorectal cancer: Results from the NursesHealth Study and Health Professionals
follow-up study. Int. J. Cancer 139, 19491957.
Kaplan, H., Thompson, P.R.C., Trumble, B.C., Samuel Wann, L., Allam, A.H., Beheim, B.,
Frohlich, P.B., Linda Sutherland, M., Sutherland, J.D., Stieglitz, J., Rodriguez, D.E.,
Michalik, D.E., Rowan, C.J., Lombardi, G.P., Bedi, R., Garcia, A.R., Min, P.J.K.,
Narula, J., Finch, P.C.E., Gurven, P.M., Thomas, P.G.S., 2017. Coronary athero-
sclerosis in indigenous South American Tsimane: a cross-sectional cohort study.
Lancet 389, 1730.
Karsdal, M.A., Krarup, H., Sand, J.M.B., Christensen, P.B., Gerstoft, J., Leeming, D.J.,
Weis, N., Schaalitzky de Muckadell, O.B., Krag, A., 2014. Review article: the e-
cacy of biomarkers in chronic broproliferative diseases - early diagnosis and prog-
nosis, with liver brosis as an exemplar. Aliment. Pharmacol. Ther. 40, 233249.
Karunadharma, P.P., Nordgaard, C.L., Olsen, T.W., Ferrington, D.A., 2010. Mitochondrial
DNA Damage as a Potential Mechanism for Age-Related Macular Degeneration.
Invest. Ophthalmol. Vis. Sci. 51, 5470.
Kasińska, M.A., Drzewoski, J., 2015. Eectiveness of probiotics in type 2 diabetes: a meta-
analysis. Pol. Arch. Med. Wewn. 125, 803813.
Katoh, A., Kai, H., Harada, H., Niiyama, H., Ikeda, H., 2017. Oral Administration of
Glucosamine Improves Vascular Endothelial Function by Modulating Intracellular
Redox State. Int. Heart J. 58, 926932.
Kaushik, S., Cuervo, A.M., 2015. Proteostasis and aging. Nat. Med. 21, 14061415.
Keefe, D., Kumar, M., Kalmbach, K., 2015. Oocyte competency is the key to embryo
potential. Fertil. Steril. 103, 317322.
Kessel, L., Siganos, G., Jørgensen, T., Larsen, M., 2011. Sleep disturbances are related to
decreased transmission of blue light to the retina caused by lens yellowing. Sleep 34,
12151219.
Khan, F., Niaz, K., Maqbool, F., Hassan, F.I., Abdollahi, M., Nagulapalli Venkata, K.C.,
Nabavi, S.M., Bishayee, A., 2016. Molecular Targets Underlying the Anticancer
Eects of Quercetin: An Update. Nutrients 8. https://doi.org/10.3390/nu8090529.
Khan, S.S., Shah, S.J., Klyachko, E., Baldridge, A.S., Eren, M., Place, A.T., Aviv, A.,
Puterman, E., Lloyd-Jones, D.M., Heiman, M., Miyata, T., Gupta, S., Shapiro, A.D.,
Vaughan, D.E., 2017. A null mutation in protects against biological aging in humans.
Sci Adv 3 eaao1617.
Khera, R., Pandey, A., Chandar, A.K., Murad, M.H., Prokop, L.J., Neeland, I.J., Berry, J.D.,
Camilleri, M., Singh, S., 2018. Eects of Weight-Loss Medications on Cardiometabolic
Risk Proles: A Systematic Review and Network Meta-analysis. Gastroenterology
154, 13091319 e7.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
20
Kikuchi, H., Inoue, S., Odagiri, Y., Inoue, M., Sawada, N., Tsugane, S., Japan Public
Health Centre (JPHC) study group, 2015. Occupational sitting time and risk of all-
cause mortality among Japanese workers. Scand. J. Work Environ. Health 41,
519528.
Kim, C.-H., Wheatley, C.M., Behnia, M., Johnson, B.D., 2016a. The Eect of Aging on
Relationships between Lean Body Mass and VO2max in Rowers. PLoS One 11,
e0160275.
Kim, C.R., Jeon, Y.-J., Kim, M.C., Jeong, T., Koo, W.R., 2018a. Reference values for hand
grip strength in the South Korean population. PLoS One 13, e0195485.
Kim, G.H., Kim, J.E., Rhie, S.J., Yoon, S., 2015a. The role of oxidative stress in neuro-
degenerative diseases. Exp. Neurobiol. 24, 325.
Kim, I.H., Kisseleva, T., Brenner, D.A., 2015b. Aging and liver disease. Curr. Opin.
Gastroenterol. 31, 184191.
Kim, G., Kim, J., Lim, Y.L., Kim, M.Y., Baik, S.K., 2016b. Renin-angiotensin system in-
hibitors and brosis in chronic liver disease: a systematic review. Hepatol. Int. 10,
819828.
Kim, K.M., Jang, H.C., Lim, S., 2016c. Dierences among skeletal muscle mass indices
derived from height-, weight-, and body mass index-adjusted models in assessing
sarcopenia. Korean J. Intern. Med. 31, 643650.
Kim, R.G., Loomba, R., Prokop, L.J., Singh, S., 2017. Statin Use and Risk of Cirrhosis and
Related Complications in Patients With Chronic Liver Diseases: A Systematic Review
and Meta-analysis. Clin. Gastroenterol. Hepatol. 15, 15211530 e8.
Kimura, Y., Hyogo, H., Yamagishi, S.-I., Takeuchi, M., Ishitobi, T., Nabeshima, Y., Arihiro,
K., Chayama, K., 2010. Atorvastatin decreases serum levels of advanced glycation
endproducts (AGEs) in nonalcoholic steatohepatitis (NASH) patients with dyslipi-
demia: clinical usefulness of AGEs as a biomarker for the attenuation of NASH. J.
Gastroenterol. 45, 750757.
Kim, Y.I., Lee, F.N., Choi, W.S., Lee, S., Youn, J.H., 2006. Insulin Regulation of Skeletal
Muscle PDK4 mRNA Expression Is Impaired in Acute Insulin-Resistant States.
Diabetes 55, 23112317.
Kim, Y.J., Kim, H.S., Seo, Y.R., 2018b. Genomic Approach to Understand the Association
of DNA Repair with Longevity and Healthy Aging Using Genomic Databases of
Oldest-Old Population. Oxid. Med. Cell. Longev., 2984730 2018.
Kim, Y., Keogh, J., Clifton, P.M., 2018c. Nuts and Cardio-Metabolic Disease: A Review of
Meta-Analyses. Nutrients 10. https://doi.org/10.3390/nu10121935.
Kiortsis, D.N., 2013. A review of the metabolic eects of controlled-release Phentermine/
Topiramate. Hormones 12, 507516.
Kirchengast, S., 2014. Physical Inactivity from the Viewpoint of Evolutionary Medicine.
Sportscience 2, 3450.
Klemera, P., Doubal, S., 2006. A new approach to the concept and computation of bio-
logical age. Mech. Ageing Dev. 127, 240248.
Knab, A.M., Lightfoot, J.T., 2010. Does the dierence between physically active and
couch potato lie in the dopamine system? Int. J. Biol. Sci. 6, 133150.
Koch, L.G., Kemi, O.J., Qi, N., Leng, S.X., Bijma, P., Gilligan, L.J., Wilkinson, J.E., Wislø,
H., Høydal, M.A., Rolim, N., Abadir, P.M., Van Grevenhof, I., Smith, G.L., Burant,
C.F., Ellingsen, Ø., Britton, S.L., Wislø, U., 2011. Intrinsic aerobic capacity sets a
divide for aging and longevity: Koch & Kemi-Rat models link exercise capacity with
mortality. Circ. Res. 109, 1162.
Koleva, D.I., Orbetzova, M.M., Atanassova, P.K., 2013. Adipose Tissue Hormones and
Appetite and Body Weight Regulators an Insulin Resistance. Folia Med. 55, 2532.
Kovsan, J., Blüher, M., Tarnovscki, T., Klöting, N., Kirshtein, B., Madar, L., Shai, I., Golan,
R., Harman-Boehm, I., Schön, M.R., Greenberg, A.S., Elazar, Z., Bashan, N., Rudich,
A., 2011. Altered autophagy in human adipose tissues in obesity. J. Clin. Endocrinol.
Metab. 96, E26877.
Kranenburg, G., Bartstra, J.W., Weijmans, M., de Jong, P.A., Mali, W.P., Verhaar, H.J.,
Visseren, F.L.J., Spiering, W., 2016. Bisphosphonates for cardiovascular risk reduc-
tion: A systematic review and meta-analysis. Atherosclerosis 252, 106115.
Kritchevsky, S.B., Beavers, K.M., Miller, M.E., Shea, M.K., Houston, D.K., Kitzman, D.W.,
Nicklas, B.J., 2015. Intentional weight loss and all-cause mortality: a meta-analysis of
randomized clinical trials. PLoS One 10, e0121993.
Krouwer, V.J.D., Hekking, L.H.P., Langelaar-Makkinje, M., Regan-Klapisz, E., Post, J.A.,
2012. Endothelial cell senescence is associated with disrupted cell-cell junctions and
increased monolayer permeability. Vasc. Cell 4, 12.
Kruk, J., Czerniak, U., 2013. Physical activity and its relation to cancer risk: updating the
evidence. Asian Pac. J. Cancer Prev. 14, 39934003.
Kundra, R., Ciryam, P., Morimoto, R.I., Dobson, C.M., Vendruscolo, M., 2017. Protein
homeostasis of a metastable subproteome associated with Alzheimers disease. Proc.
Natl. Acad. Sci. U. S. A. 114, E5703E5711.
Kurata, N., Schachern, P.A., Paparella, M.M., Cureoglu, S., 2016. Histopathologic
Evaluation of Vascular Findings in the Cochlea in Patients With Presbycusis. JAMA
Otolaryngol. Head Neck Surg. 142, 173178.
Kusy, K., Zieliński, J., 2014. Aerobic capacity in speed-power athletes aged 20-90 years vs
endurance runners and untrained participants. Scand. J. Med. Sci. Sports 24, 6879.
Lakatta, E.G., 2015. So! Whats aging? Is cardiovascular aging a disease? J. Mol. Cell.
Cardiol. 83, 113.
Langmann, G.A., Perera, S., Ferchak, M.A., Nace, D.A., Resnick, N.M., Greenspan, S.L.,
2017. Inammatory Markers and Frailty in Long-Term Care Residents. J. Am. Geriatr.
Soc. 65, 17771783.
Lan, J., Zhao, Y., Dong, F., Yan, Z., Zheng, W., Fan, J., Sun, G., 2015a. Meta-analysis of
the eect and safety of berberine in the treatment of type 2 diabetes mellitus, hy-
perlipemia and hypertension. J. Ethnopharmacol. 161, 6981.
Lan, J., Zhao, Y., Dong, F., Yan, Z., Zheng, W., Fan, J., Sun, G., 2015b. Meta-analysis of
the eect and safety of berberine in the treatment of type 2 diabetes mellitus, hy-
perlipemia and hypertension. J. Ethnopharmacol. 161, 6981.
Lark, D.S., Wasserman, D.H., 2017. Meta-brosis links positive energy balance and mi-
tochondrial metabolism to insulin resistance. F1000Res 6. https://doi.org/10.12688/
f1000research.11653.1.
Lee, D.M., Tajar, A., Ravindrarajah, R., Pye, S.R., OConnor, D.B., Corona, G., OConnell,
M., Gielen, E., Boonen, S., Vanderschueren, D., Pendleton, N., Finn, J.D., Bartfai, G.,
Casanueva, F.F., Forti, G., Giwercman, A., Han, T.S., Huhtaniemi, I.T., Kula, K., Lean,
M.E.J., Punab, M., Wu, F.C.W., ONeill, T.W., the European Male Aging Study Group,
2012. Frailty and Sexual Health in Older European Men. J. Gerontol. A Biol. Sci. Med.
Sci. 68, 837844.
Lee, S., Paudel, O., Jiang, Y., Yang, X.-R., Sham, J.S.K., 2015. CD38 mediates angiotensin
II-induced intracellular Ca(2+) release in rat pulmonary arterial smooth muscle cells.
Am. J. Respir. Cell Mol. Biol. 52, 332341.
Lehagen, U., Aaseth, J., Alexander, J., Svensson, E., Johansson, P., Larsson, A., 2018. Less
brosis in elderly subjects supplemented with selenium and coenzyme Q10-A me-
chanism behind reduced cardiovascular mortality? BioFactors 44, 137147.
Lehallier, B., Gate, D., Schaum, N., Nanasi, T., Lee, S.E., Yousef, H., Moran Losada, P.,
Berdnik, D., Keller, A., Verghese, J., Sathyan, S., Franceschi, C., Milman, S., Barzilai,
N., Wyss-Coray, T., 2019. Undulating changes in human plasma proteome proles
across the lifespan. Nat. Med. 25, 18431850.
Lei, W.-T., Shih, P.-C., Liu, S.-J., Lin, C.-Y., Yeh, T.-L., 2017. Eect of Probiotics and
Prebiotics on Immune Response to Inuenza Vaccination in Adults: A Systematic
Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 9. https://doi.
org/10.3390/nu9111175.
Lei, X., Yu, J., Niu, Q., Liu, J., Fraering, P.C., Wu, F., 2015. The FDA-approved natural
product dihydroergocristine reduces the production of the Alzheimers disease
amyloid-βpeptides. Sci. Rep. 5. https://doi.org/10.1038/srep16541.
Lemogoum, D., Ngatchou, W., Janssen, C., Leeman, M., Van Bortel, L., Boutouyrie, P.,
Degaute, J.P., Van de Borne, P., 2012. Eects of hunter-gatherer subsistence mode on
arterial distensibility in Cameroonian pygmies. Hypertension 60, 123128.
Levine, M.E., Lu, A.T., Quach, A., Chen, B.H., Assimes, T.L., Bandinelli, S., Hou, L.,
Baccarelli, A.A., Stewart, J.D., Li, Y., Whitsel, E.A., Wilson, J.G., Reiner, A.P., Aviv,
A., Lohman, K., Liu, Y., Ferrucci, L., Horvath, S., 2018. An epigenetic biomarker of
aging for lifespan and healthspan. Aging 10, 573591.
Liang, H., Tantiwong, P., Sriwijitkamol, A., Shanmugasundaram, K., Mohan, S., Espinoza,
S., DeFronzo, R.A., Dubé, J.J., Musi, N., 2013. Eect of a sustained reduction in
plasma free fatty acid concentration on insulin signalling and inammation in ske-
letal muscle from human subjects. J. Physiol. 591, 2897.
Liao, Y., Dong, Y., Cheng, J., 2017. The Function of the Mitochondrial Calcium Uniporter
in Neurodegenerative Disorders. Int. J. Mol. Sci. 18, 248.
Li, G., Liu, K.-Y., Qiu, Z.-P., 2019. An integrative module analysis of DNA methylation
landscape in aging. Exp. Ther. Med. 17, 34113416.
Li, G., Mayer, C.L., Morelli, D., Millard, S.P., Raskind, W.H., Petrie, E.C., Cherrier, M.,
Fagan, A.M., Raskind, M.A., Peskind, E.R., 2017a. Eect of simvastatin on CSF
Alzheimer disease biomarkers in cognitively normal adults. Neurology 89,
12511255.
Li, H., Liu, W., Xie, J., 2017b. Circulating interleukin-6 levels and cardiovascular and all-
cause mortality in the elderly population: A meta-analysis. Arch. Gerontol. Geriatr.
73, 257262.
Li, L., Yan, L., 2017. Ecacy of coenzyme Q10 in patients with cardiac failure: a meta-
analysis of clinical trials. BMC Cardiovasc. Disord. 17, 196.
Lim, Y.-A., Rhein, V., Baysang, G., Meier, F., Poljak, A., Raftery, M.J., Guilhaus, M., Ittner,
L.M., Eckert, A., Götz, J., 2010. Abeta and human amylin share a common toxicity
pathway via mitochondrial dysfunction. Proteomics 10, 16211633.
Lindqvist, P.G., Landin-Olsson, M., 2017. The relationship between sun exposure and all-
cause mortality. Photochem. Photobiol. Sci. 16, 354361.
Lin, H.-S., Watts, J.N., Peel, N.M., Hubbard, R.E., 2016. Frailty and post-operative out-
comes in older surgical patients: a systematic review. BMC Geriatr. 16, 157.
Lin, X., Chen, W., Wei, F., Ying, M., Wei, W., Xie, X., 2015. Night-shift work increases
morbidity of breast cancer and all-cause mortality: a meta-analysis of 16 prospective
cohort studies. Sleep Med. 16, 13811387.
Lipson, K.E., Wong, C., Teng, Y., Spong, S., 2012. CTGF is a central mediator of tissue
remodeling and brosis and its inhibition can reverse the process of brosis.
Fibrogenesis Tissue Repair 5, S24.
Li, S., Li, Y., Xiang, L., Dong, J., Liu, M., Xiang, G., 2018. Sildenal induces browning of
subcutaneous white adipose tissue in overweight adults. Metabolism 78, 106117.
Li, S., Xu, M., Niu, Q., Xu, S., Ding, Y., Yan, Y., Guo, S., Li, F., 2015. Ecacy of
Procyanidins against In Vivo Cellular Oxidative Damage: A Systematic Review and
Meta-Analysis. PLoS One 10, e0139455.
Liton, P.B., Challa, P., Stinnett, S., Luna, C., Epstein, D.L., Gonzalez, P., 2005. Cellular
senescence in the glaucomatous outow pathway. Exp. Gerontol. 40, 745748.
Liu, F., Wang, X., Wu, G., Chen, L., Hu, P., Ren, H., Hu, H., 2015. Coee Consumption
Decreases Risks for Hepatic Fibrosis and Cirrhosis: A Meta-Analysis. PLoS One 10,
e0142457.
Liu, J., Zeng, F.-F., Liu, Z.-M., Zhang, C.-X., Ling, W.-H., Chen, Y.-M., 2013. Eects of
blood triglycerides on cardiovascular and all-cause mortality: a systematic review
and meta-analysis of 61 prospective studies. Lipids Health Dis. 12, 159.
Liu, Y., Sano, H.K., Cho, H., Burd, C.E., Torrice, C., Ibrahim, J.G., Thomas, N.E.,
Sharpless, N.E., 2009. Expression ofp16INK4ain peripheral blood T-cells is a bio-
marker of human aging. Aging Cell 8, 439448.
Li, X., Zhu, L., Wang, B., Yuan, M., Zhu, R., 2017c. Drugs and Targets in Fibrosis. Front.
Pharmacol. 8, 855.
Li, Y.-S., Zhang, F.-J., Zeng, C., Luo, W., Xiao, W.-F., Gao, S.-G., Lei, G.-H., 2016.
Autophagy in osteoarthritis. Joint Bone Spine 83, 143148.
Lobo, R.A., 2017. Hormone-replacement therapy: current thinking. Nat. Rev. Endocrinol.
13, 220231.
Lockyer, S., Rowland, I., Spencer, J.P.E., Yaqoob, P., Stonehouse, W., 2017. Impact of
phenolic-rich olive leaf extract on blood pressure, plasma lipids and inammatory
markers: a randomised controlled trial. Eur. J. Nutr. 56, 1421.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
21
Loguercio, C., Nardi, G., Argenzio, F., Aurilio, C., Petrone, E., Grella, A., Del Vecchio
Blanco, C., Coltorti, M., 1994. Eect of S-adenosyl-L-methionine administration on
red blood cell cysteine and glutathione levels in alcoholic patients with and without
liver disease. Alcohol Alcohol 29, 597604.
López-Otín, C., Blasco, M.A., Partridge, L., Serrano, M., Kroemer, G., 2013. The hallmarks
of aging. Cell 153, 11941217.
Lorenzatti, A.J., Retzla, B.M., 2016. Unmet needs in the management of atherosclerotic
cardiovascular disease: Is there a role for emerging anti-inammatory interventions?
Int. J. Cardiol. 221, 581586.
Lowery, E.M., Brubaker, A.L., Kuhlmann, E., Kovacs, E.J., 2013. The aging lung. Clin.
Interv. Aging 14891496.
Lu, A.T., Quach, A., Wilson, J.G., Reiner, A.P., Aviv, A., Raj, K., Hou, L., Baccarelli, A.A.,
Li, Y., Stewart, J.D., Whitsel, E.A., Assimes, T.L., Ferrucci, L., Horvath, S., 2019. DNA
methylation GrimAge strongly predicts lifespan and healthspan. Aging 11, 303327.
Lucia Pagani, A.E., 2011. Amyloid-Beta Interaction with Mitochondria. Int. J. Alzheimers.
Dis. https://doi.org/10.4061/2011/925050. 2011.
Lu, F.-P., Lin, K.-P., Kuo, H.-K., 2009. Diabetes and the risk of multi-system aging phe-
notypes: a systematic review and meta-analysis. PLoS One 4, e4144.
Lv, W., Du, N., Liu, Y., Fan, X., Wang, Y., Jia, X., Hou, X., Wang, B., 2016. Low
Testosterone Level and Risk of Alzheimers Disease in the Elderly Men: a Systematic
Review and Meta-Analysis. Mol. Neurobiol. 53, 26792684.
Ly, H., Despa, F., 2015. Hyperamylinemia as a risk factor for accelerated cognitive decline
in diabetes. Expert Rev. Proteomics 12, 575.
Maharlouei, N., Tabrizi, R., Lankarani, K.B., Rezaianzadeh, A., Akbari, M., Kolahdooz, F.,
Rahimi, M., Keneshlou, F., Asemi, Z., 2018. The eects of ginger intake on weight loss
and metabolic proles amng overweight and obese subjects: A systematic review and
meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 114.
Makimura, H., Stanley, T.L., Suresh, C., De Sousa-Coelho, A.L., Frontera, W.R., Syu, S.,
Braun, L.R., Looby, S.E., Feldpausch, M.N., Torriani, M., Lee, H., Patti, M.-E.,
Grinspoon, S.K., 2016. Metabolic Eects of Long-Term Reduction in Free Fatty Acids
With Acipimox in Obesity: A Randomized Trial. J. Clin. Endocrinol. Metab. 101,
1123.
Malambo, P., Kengne, A.P., De Villiers, A., Lambert, E.V., Puoane, T., 2016. Built
Environment, Selected Risk Factors and Major Cardiovascular Disease Outcomes: A
Systematic Review. PLoS One 11. https://doi.org/10.1371/journal.pone.0166846.
Mamoshina, P., Kochetov, K., Cortese, F., Kovalchuk, A., Aliper, A., Putin, E., Scheibye-
Knudsen, M., Cantor, C.R., Skjodt, N.M., Kovalchuk, O., Zhavoronkov, A., 2019.
Blood Biochemistry Analysis to Detect Smoking Status and Quantify Accelerated
Aging in Smokers. Sci. Rep. 9, 142.
Mamoshina, P., Kochetov, K., Putin, E., Cortese, F., Aliper, A., Lee, W.-S., Ahn, S.-M., Uhn,
L., Skjodt, N., Kovalchuk, O., Scheibye-Knudsen, M., Zhavoronkov, A., 2018.
Population Specic Biomarkers of Human Aging: A Big Data Study Using South
Korean, Canadian, and Eastern European Patient Populations. J. Gerontol. A Biol. Sci.
Med. Sci. 73, 14821490.
Mancini, J.G., Filion, K.B., Atallah, R., Eisenberg, M.J., 2016. Systematic Review of the
Mediterranean Diet for Long-Term Weight Loss. Am. J. Med. 129, 407415 e4.
Manini, T.M., 2010. Energy expenditure and aging. Ageing Res. Rev. 9, 111.
Mannick, J.B., Del Giudice, G., Lattanzi, M., Valiante, N.M., Praestgaard, J., Huang, B.,
Lonetto, M.A., Maecker, H.T., Kovarik, J., Carson, S., Glass, D.J., Klickstein, L.B.,
2014. mTOR inhibition improves immune function in the elderly. Sci. Transl. Med. 6,
268ra179.
Marjoribanks, J., Farquhar, C., Roberts, H., Lethaby, A., Lee, J., 2017. Long-term hormone
therapy for perimenopausal and postmenopausal women. Cochrane Database Syst.
Rev. 1 CD004143.
Martini, D., Del Bo, C., Tassotti, M., Riso, P., Del Rio, D., Brighenti, F., Porrini, M., 2016.
Coee Consumption and Oxidative Stress: A Review of Human Intervention Studies.
Molecules 21. https://doi.org/10.3390/molecules21080979.
Marty, F.M., Ljungman, P., Chemaly, R.F., Maertens, J., Dadwal, S.S., Duarte, R.F.,
Haider, S., Ullmann, A.J., Katayama, Y., Brown, J., Mullane, K.M., Boeckh, M.,
Blumberg, E.A., Einsele, H., Snydman, D.R., Kanda, Y., DiNubile, M.J., Teal, V.L.,
Wan, H., Murata, Y., Kartsonis, N.A., Leavitt, R.Y., Badshah, C., 2017. Letermovir
Prophylaxis for Cytomegalovirus in Hematopoietic-Cell Transplantation. N. Engl. J.
Med. 377, 24332444.
Marventano, S., Vetrani, C., Vitale, M., Godos, J., Riccardi, G., Grosso, G., 2017. Whole
Grain Intake and Glycaemic Control in Healthy Subjects: A Systematic Review and
Meta-Analysis of Randomized Controlled Trials. Nutrients 9. https://doi.org/10.
3390/nu9070769.
Marzetti, E., Calvani, R., Cesari, M., Buford, T.W., Lorenzi, M., Behnke, B.J.,
Leeuwenburgh, C., 2013. Mitochondrial dysfunction and sarcopenia of aging: from
signaling pathways to clinical trials. Int. J. Biochem. Cell Biol. 45, 2288.
Masarone, M., Rosato, V., Dallio, M., Gravina, A.G., Aglitti, A., Loguercio, C., Federico, A.,
Persico, M., 2018. Role of Oxidative Stress in Pathophysiology of Nonalcoholic Fatty
Liver Disease. Oxid. Med. Cell. Longev., 9547613 2018.
Massudi, H., Grant, R., Braidy, N., Guest, J., Farnsworth, B., Guillemin, G.J., 2012. Age-
associated changes in oxidative stress and NAD+ metabolism in human tissue. PLoS
One 7, e42357.
Mathers, J.C., Deary, I.J., Kuh, D., Lord, J.M., Khaw, K.-T., Lara, J., Nissan, J., Cooper, R.,
Ginty, A., 2012. Guidelines for biomarkers of healthy ageing. Medical Research
Council, Swindon.
Maurizi, G., Della Guardia, L., Maurizi, A., Poloni, A., 2018. Adipocytes properties and
crosstalk with immune system in obesity-related inammation. J. Cell. Physiol. 233,
8897.
Mazidi, M., Gao, H.-K., Rezaie, P., Ferns, G.A., 2016. The eect of ginger supplementation
on serum C-reactive protein, lipid prole and glycaemia: a systematic review and
meta-analysis. Food Nutr. Res. 60. https://doi.org/10.3402/fnr.v60.32613.
Mazidi, M., Rezaie, P., Ferns, G.A., Vatanparast, H., 2017. Impact of Probiotic
Administration on Serum C-Reactive Protein Concentrations: Systematic Review and
Meta-Analysis of Randomized Control Trials. Nutrients 9. https://doi.org/10.3390/
nu9010020.
McCarthy, M.M., Hannan, J.A., 2014. The Mature Athlete: Aging Tendon and Ligament.
Sports Health 6, 4148.
McCarty, M.F., OKeefe, J.H., DiNicolantonio, J.J., 2016. Pentoxifylline for vascular
health: a brief review of the literature. Open Heart 3. https://doi.org/10.1136/
openhrt-2015-000365.
McCulloch, K., Litherland, G.J., Rai, T.S., 2017. Cellular senescence in osteoarthritis
pathology. Aging Cell 16, 210218.
McCullough, A., 2015. Alternatives to testosterone replacement: testosterone restoration.
Asian J. Androl. 17, 201205.
McLoughlin, R.F., Berthon, B.S., Jensen, M.E., Baines, K.J., Wood, L.G., 2017. Short-chain
fatty acids, prebiotics, synbiotics, and systemic inammation: a systematic review
and meta-analysis. Am. J. Clin. Nutr. 106, 930945.
Mei, Z., Liang, M., Li, L., Zhang, Y., Wang, Q., Yang, W., 2017. Eects of statins on cancer
mortality and progression: A systematic review and meta-analysis of 95 cohorts in-
cluding 1,111,407 individuals. Int. J. Cancer 140, 10681081.
Menezes, R., Rodriguez-Mateos, A., Kaltsatou, A., González-Sarrías, A., Greyling, A.,
Giannaki, C., Andres-Lacueva, C., Milenkovic, D., Gibney, E.R., Dumont, J., Schär, M.,
Garcia-Aloy, M., Palma-Duran, S.A., Ruskovska, T., Maksimova, V., Combet, E., Pinto,
P., 2017. Impact of Flavonols on Cardiometabolic Biomarkers: A Meta-Analysis of
Randomized Controlled Human Trials to Explore the Role of Inter-Individual
Variability. Nutrients 9. https://doi.org/10.3390/nu9020117.
Meng, X.-M., Nikolic-Paterson, D.J., Lan, H.Y., 2016. TGF-β: the master regulator of -
brosis. Nat. Rev. Nephrol. 12, 325338.
Menicacci, B., Cipriani, C., Margheri, F., Mocali, A., Giovannelli, L., 2017. Modulation of
the Senescence-Associated Inammatory Phenotype in Human Fibroblasts by Olive
Phenols. Int. J. Mol. Sci. 18. https://doi.org/10.3390/ijms18112275.
Menon, K.A., Mousa, A., de Courten, B., 2018. Eect of Carnosine Supplementation on
Cardiometabolic Risk Factors in Obesity, Prediabetes, and DiabetesA Meta-analysis
of Randomized Controlled Trials. Diabetes 67 55LB.
Meslé, F., Vallin, J., 2017. Causes of death at very old age, including supercentenarians.
In: Presented at the PAA meeting. French Institute for Demographic Studies (INED).
Miao, J., Manthena, P.V., Haas, M.E., Ling, A.V., Shin, D.-J., Graham, M.J., Crooke, R.M.,
Liu, J., Biddinger, S.B., 2015. The Role of Insulin in the Regulation of PCSK9.
Arterioscler. Thromb. Vasc. Biol. 35, 1589.
Mirhosseini, N., Rainsbury, J., Kimball, S.M., 2018. Vitamin D Supplementation, Serum
25(OH)D Concentrations and Cardiovascular Disease Risk Factors: A Systematic
Review and Meta-Analysis. Front Cardiovasc Med. 5, 87.
Mitnitski, A.B., Mogilner, A.J., Rockwood, K., 2001. Accumulation of decits as a proxy
measure of aging. ScienticWorldJournal 1, 323336.
Mitnitski, A., Collerton, J., Martin-Ruiz, C., Jagger, C., von Zglinicki, T., Rockwood, K.,
Kirkwood, T.B.L., 2015. Age-related frailty and its association with biological mar-
kers of ageing. BMC Med. 13, 161.
Moeller, M., Pink, C., Endlich, N., Endlich, K., Grabe, H.-J., Völzke, H., Dörr, M., Nauck,
M., Lerch, M.M., Köhling, R., Holtfreter, B., Kocher, T., Fuellen, G., 2017. Mortality is
associated with inammation, anemia, specic diseases and treatments, and mole-
cular markers. PLoS One 12, e0175909.
Mohammadi-Sartang, M., Mazloom, Z., Sherafatmanesh, S., Ghorbani, M., Firoozi, D.,
2017. Eects of supplementation with quercetin on plasma C-reactive protein con-
centrations: a systematic review and meta-analysis of randomized controlled trials.
Eur. J. Clin. Nutr. 71, 10331039.
Mons, U., Müezzinler, A., Gellert, C., Schöttker, B., Abnet, C.C., Bobak, M., de Groot, L.,
Freedman, N.D., Jansen, E., Kee, F., Kromhout, D., Kuulasmaa, K., Laatikainen, T.,
ODoherty, M.G., Bueno-de-Mesquita, B., Orfanos, P., Peters, A., van der Schouw,
Y.T., Wilsgaard, T., Wolk, A., Trichopoulou, A., Boetta, P., Brenner, H., CHANCES
Consortium, 2015. Impact of smoking and smoking cessation on cardiovascular
events and mortality among older adults: meta-analysis of individual participant data
from prospective cohort studies of the CHANCES consortium. BMJ 350, h1551.
Monteiro-Junior, R.S., de Tarso Maciel-Pinheiro, P., da Matta Mello Portugal, E., da Silva
Figueiredo, L.F., Terra, R., Carneiro, L.S.F., Rodrigues, V.D., Nascimento, O.J.M.,
Deslandes, A.C., Laks, J., 2018. Eect of Exercise on Inammatory Prole of Older
Persons: Systematic Review and Meta-Analyses. J Phys Act Health 15, 6471.
Mookerjee, S.A., Divakaruni, A.S., Jastroch, M., Brand, M.D., 2010. Mitochondrial un-
coupling and lifespan. Mech. Ageing Dev. 131, 463.
Mora Huertas, A.C., Schmelzer, C.E.H., Hoehenwarter, W., Heyroth, F., Heinz, A., 2016.
Molecular-level insights into aging processes of skin elastin. Biochimie 128-129,
163173.
Morimoto, R.I., Cuervo, A.M., 2014. Proteostasis and the aging proteome in health and
disease. J. Gerontol. A Biol. Sci. Med. Sci. 69 (Suppl 1), S338.
Moro-García, M.A., López Iglesias, F., Avanzas, P., Echeverría, A., López-Larrea, C., Morís
de la Tassa, C., Alonso-Arias, R., 2015. Disease complexity in acute coronary syn-
drome is related to the patients immunological status. Int. J. Cardiol. 189, 115123.
Morry, J., Ngamcherdtrakul, W., Yantasee, W., 2017. Oxidative stress in cancer and -
brosis: Opportunity for therapeutic intervention with antioxidant compounds, en-
zymes, and nanoparticles. Redox Biology 11, 240.
Mosconi, L., Sorbi, S., de Leon, M.J., Li, Y., Nacmias, B., Myoung, P.S., Tsui, W.,
Ginestroni, A., Bessi, V., Fayyazz, M., Caarra, P., Pupi, A., 2006. Hypometabolism
exceeds atrophy in presymptomatic early-onset familial Alzheimers disease. J. Nucl.
Med. 47, 17781786.
Moskalev, A., Chernyagina, E., Kudryavtseva, A., Shaposhnikov, M., 2017.
Geroprotectors: A Unied Concept and Screening Approaches. Aging Dis. 8, 354.
Mousavi, S.M., Milajerdi, A., Kord, V.H., Mohsen, G.M., Esmaillzadeh, A., 2018a. The
eects of curcumin supplementation on body weight, body mass index and waist
circumference: a systematic review and dose-response meta-analysis of randomized
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
22
controlled trials. Crit. Rev. Food Sci. Nutr. 110.
Mousavi, S.M., Milajerdi, A., Sheikhi, A., Kord-Varkaneh, H., Feinle-Bisset, C., Larijani, B.,
Esmaillzadeh, A., 2018b. Resveratrol supplementation signicantly inuences obe-
sity measures: a systematic review and dose-response meta-analysis of randomized
controlled trials. Obes. Rev. https://doi.org/10.1111/obr.12775.
Mou, X., Zhou, D.-Y., Zhou, D.-Y., Ma, J.-R., Liu, Y.-H., Chen, H.-P., Hu, Y.-B., Shou, C.-M.,
Chen, J.-W., Liu, W.-H., Ma, G.-L., 2016. Serum TGF-β1 as a Biomarker for Type 2
Diabetic Nephropathy: A Meta-Analysis of Randomized Controlled Trials. PLoS One
11, e0149513.
Mundstock, E., Zatti, H., Louzada, F.M., Oliveira, S.G., Guma, F.T.C.R., Paris, M.M.,
Rueda, A.B., Machado, D.G., Stein, R.T., Jones, M.H., Sarria, E.E., Barbé-Tuana, F.M.,
Mattiello, R., 2015. Eects of physical activity in telomere length: Systematic review
and meta-analysis. Ageing Res. Rev. 22, 7280.
Muñoz, A.M., Falque-Madrid, L., Zambrano, R.C., Maestre, G.E., 2010. Basic anthro-
pometry and health status of elderly: ndings of the Maracaibo Aging Study. J. Aging
Health 22, 242261.
Murray, M.A., Chotirmall, S.H., 2015. The Impact of Immunosenescence on Pulmonary
Disease. Mediators Inamm., 692546 2015.
Naci, H., Brugts, J.J., Fleurence, R., Tsoi, B., Toor, H., Ades, A.E., 2013. Comparative
benets of statins in the primary and secondary prevention of major coronary events
and all-cause mortality: a network meta-analysis of placebo-controlled and active-
comparator trials. Eur. J. Prev. Cardiol. 20, 641657.
Naci, H., Ioannidis, J.P.A., 2013. Comparative eectiveness of exercise and drug inter-
ventions on mortality outcomes: metaepidemiological study. BMJ 347, f5577.
Nagpal, R., Mainali, R., Ahmadi, S., Wang, S., Singh, R., Kavanagh, K., Kitzman, D.W.,
Kushugulova, A., Marotta, F., Yadav, H., 2018. Gut microbiome and aging:
Physiological and mechanistic insights. Nutr Healthy Aging 4, 267285.
Nakamura, T., Sato, E., Fujiwara, N., Kawagoe, Y., Takeuchi, M., Maeda, S., Yamagishi,
S.-I., 2010. Atorvastatin reduces proteinuria in non-diabetic chronic kidney disease
patients partly via lowering serum levels of advanced glycation end products (AGEs).
Oxid. Med. Cell. Longev. 3, 304307.
Nascimento, C.M.C., Zazzetta, M.S., Gomes, G.A.O., Orlandi, F.S., Gramani-Say, K.,
Vasilceac, F.A., Gratão, A.C.M., Pavarini, S.C.I., Cominetti, M.R., 2018. Higher levels
of tumor necrosis factor βare associated with frailty in socially vulnerable commu-
nity-dwelling older adults. BMC Geriatr. 18, 268.
Navarro, S.L., White, E., Kantor, E.D., Zhang, Y., Rho, J., Song, X., Milne, G.L., Lampe,
P.D., Lampe, J.W., 2015. Randomized trial of glucosamine and chondroitin supple-
mentation on inammation and oxidative stress biomarkers and plasma proteomics
proles in healthy humans. PLoS One 10, e0117534.
Neale, E.P., Tapsell, L.C., Guan, V., Batterham, M.J., 2017. The eect of nut consumption
on markers of inammation and endothelial function: a systematic review and meta-
analysis of randomised controlled trials. BMJ Open 7, e016863.
Nenna, A., Spadaccio, C., Lusini, M., Ulianich, L., Chello, M., Nappi, F., 2015. Basic and
Clinical Research Against Advanced Glycation End Products (AGEs): New
Compounds to Tackle Cardiovascular Disease and Diabetic Complications. Recent
Adv Cardiovasc Drug Discov 10, 1033.
Neviere, R., Yu, Y., Wang, L., Tessier, F., Boulanger, E., 2016. Implication of advanced
glycation end products (Ages) and their receptor (Rage) on myocardial contractile
and mitochondrial functions. Glycoconj. J. 33, 607617.
Nieman, D.C., Williams, A.S., Shanely, R.A., Jin, F., McAnulty, S.R., Triplett, N.T., Austin,
M.D., Henson, D.A., 2010. Quercetinsinuence on exercise performance and muscle
mitochondrial biogenesis. Med. Sci. Sports Exerc. 42, 338345.
Nisoli, E., Tonello, C., Cardile, A., Cozzi, V., Bracale, R., Tedesco, L., Falcone, S., Valerio,
A., Cantoni, O., Clementi, E., Moncada, S., Carruba, M.O., 2005. Calorie restriction
promotes mitochondrial biogenesis by inducing the expression of eNOS. Science 310,
314317.
North, B.J., Sinclair, D.A., 2012. The intersection between aging and cardiovascular
disease. Circ. Res. 110, 10971108.
Nygaard, M., Thinggaard, M., Christensen, K., Christiansen, L., 2017. Investigation of the
5q33.3 longevity locus and age-related phenotypes. Aging 9, 247255.
Ojo, J.O., Rezaie, P., Gabbott, P.L., Stewart, M.G., 2015. Impact of age-related neuroglial
cell responses on hippocampal deterioration. Front. Aging Neurosci. 7. https://doi.
org/10.3389/fnagi.2015.00057.
Okada, M., Yamashita, S., Ueyama, H., Ishizaki, M., Maeda, Y., Ando, Y., 2018. Long-term
eects of edaravone on survival of patients with amyotrophic lateral sclerosis.
eNeurologicalSci 11, 11.
Oktay, K., Turan, V., Titus, S., Stobezki, R., Liu, L., 2015. BRCA Mutations, DNA Repair
Deciency, and Ovarian Aging. Biol. Reprod. 93, 67.
Ommundsen, N., Wyller, T.B., Nesbakken, A., Jordhøy, M.S., Bakka, A., Skovlund, E.,
Rostoft, S., 2014. Frailty is an independent predictor of survival in older patients with
colorectal cancer. Oncologist 19, 12681275.
Ooms, S., Overeem, S., Besse, K., Rikkert, M.O., Verbeek, M., Claassen, J.A.H.R., 2014.
Eect of 1 night of total sleep deprivation on cerebrospinal uid β-amyloid 42 in
healthy middle-aged men: a randomized clinical trial. JAMA Neurol. 71, 971977.
Palmeri, A., Ricciarelli, R., Gulisano, W., Rivera, D., Rebosio, C., Calcagno, E., Tropea,
M.R., Conti, S., Das, U., Roy, S., Pronzato, M.A., Arancio, O., Fedele, E., Puzzo, D.,
2017. Amyloid-βPeptide Is Needed for cGMP-Induced Long-Term Potentiation and
Memory. J. Neurosci. 37, 6926.
Pal, S., Tyler, J.K., 2016. Epigenetics and aging. Science Advances 2. https://doi.org/10.
1126/sciadv.1600584.
Pan, A., Yu, D., Demark-Wahnefried, W., Franco, O.H., Lin, X., 2009. Meta-analysis of the
eects of axseed interventions on blood lipids. Am. J. Clin. Nutr. 90, 288297.
Paradisi, G., Bracaglia, M., Basile, F., DIppolito, S., Di Nicuolo, F., Ianniello, F.,
Quagliozzi, L., Donati, L., Labianca, A., Di Cesare, C., Viggiano, M., Biaggi, A., De
Waure, C., Andreotti, F., Di Simone, N., Caruso, A., 2012. Eect of pravastatin on
endothelial function and endothelial progenitor cells in healthy postmenopausal
women. Clinical and Experimental Obstetrics and Gynecology 39, 153159.
Pararasa, C., Bailey, C.J., Griths, H.R., 2015. Ageing, adipose tissue, fatty acids and
inammation. Biogerontology 16, 235248.
Parker, L., Caldow, M.K., Watts, R., Levinger, P., Cameron-Smith, D., Levinger, I., 2017.
Age and sex dierences in human skeletal muscle brosis markers and transforming
growth factor-βsignaling. Eur. J. Appl. Physiol. 117, 14631472.
Park, M.H., Kim, D.H., Lee, E.K., Kim, N.D., Im, D.S., Lee, J., Yu, B.P., Chung, H.Y., 2014.
Age-related inammation and insulin resistance: a review of their intricate inter-
dependency. Arch. Pharm. Res. 37, 1507.
Pasupathy, S., Tavella, R., Grover, S., Raman, B., Procter, N.E.K., Du, Y.T., Mahadavan,
G., Staord, I., Heresztyn, T., Holmes, A., Zeitz, C., Arstall, M., Selvanayagam, J.,
Horowitz, J.D., Beltrame, J.F., 2017. Early Use of N-acetylcysteine With Nitrate
Therapy in Patients Undergoing Primary Percutaneous Coronary Intervention for ST-
Segment-Elevation Myocardial Infarction Reduces Myocardial Infarct Size (the
NACIAM Trial [N-acetylcysteine in Acute Myocardial Infarction]). Circulation 136,
894903.
Pavlidis, P., Gouveris, H., Anogeianaki, A., Koutsonikolas, D., Anogianakis, G., Kekes, G.,
2013. Age-related changes in electrogustometry thresholds, tongue tip vasculariza-
tion, density, and form of the fungiform papillae in humans. Chem. Senses 38, 3543.
Payne, B.A.I., Chinnery, P.F., 2015. Mitochondrial dysfunction in aging: Much progress
but many unresolved questions. Biochim. Biophys. Acta 1847, 1347.
Pchitskaya, E., Popugaeva, E., Bezprozvanny, I., 2018. Calcium signaling and molecular
mechanisms underlying neurodegenerative diseases. Cell Calcium 70, 8794.
Pelucchi, C., Bosetti, C., Negri, E., Lipworth, L., La Vecchia, C., 2011. Olive oil and cancer
risk: an update of epidemiological ndings through 2010. Curr. Pharm. Des. 17,
805812.
Pera, A., Campos, C., López, N., Hassouneh, F., Alonso, C., Tarazona, R., Solana, R., 2015.
Immunosenescence: Implications for response to infection and vaccination in older
people. Maturitas 82, 5055.
Perumpail, B.J., Cholankeril, R., Yoo, E.R., Kim, D., Ahmed, A., 2017. An Overview of
Dietary Interventions and Strategies to Optimize the Management of Non-Alcoholic
Fatty Liver Disease. Diseases 5, 23.
Pescosolido, N., Barbato, A., Giannotti, R., Komaiha, C., Lenarduzzi, F., 2016. Age-related
changes in the kinetics of human lenses: prevention of the cataract. Int. J.
Ophthalmol. 9, 1506.
Pes, G.M., Tolu, F., Poulain, M., Errigo, A., Masala, S., Pietrobelli, A., Battistini, N.C.,
Maioli, M., 2013. Lifestyle and nutrition related to male longevity in Sardinia: an
ecological study. Nutr. Metab. Cardiovasc. Dis. 23, 212219.
Peterson, C.M., Johannsen, D.L., Ravussin, E., 2012. Skeletal muscle mitochondria and
aging: a review. J. Aging Res., 194821 2012.
Picca, A., Lezza, A.M.S., Leeuwenburgh, C., Pesce, V., Calvani, R., Bossola, M., Manes-
Gravina, E., Landi, F., Bernabei, R., Marzetti, E., 2018. Circulating Mitochondrial
DNA at the Crossroads of Mitochondrial Dysfunction and Inammation During Aging
and Muscle Wasting Disorders. Rejuvenation Res. 21, 350359.
Piera-Velazquez, S., Mendoza, F.A., Jimenez, S.A., 2016. Endothelial to Mesenchymal
Transition (EndoMT) in the Pathogenesis of Human Fibrotic Diseases. J. Clin. Med.
Res. 5. https://doi.org/10.3390/jcm5040045.
Pollice, P.F., Rosier, R.N., Looney, R.J., Puzas, J.E., Schwarz, E.M., OKeefe, R.J., 2001.
Oral pentoxifylline inhibits release of tumor necrosis factor-alpha from human per-
ipheral blood monocytes : a potential treatment for aseptic loosening of total joint
components. J. Bone Joint Surg. Am. 83-A, 10571061.
Poole, R., Kennedy, O.J., Roderick, P., Falloweld, J.A., Hayes, P.C., Parkes, J., 2017.
Coee consumption and health: umbrella review of meta-analyses of multiple health
outcomes. BMJ 359, j5024.
Poolsup, N., Suksomboon, N., Plordplong, N., 2016. Eect of vitamin D supplementation
on insulin resistance and glycaemic control in prediabetes: a systematic review and
meta-analysis. Diabet. Med. 33, 290299.
Pope 3rd, C.A., Coleman, N., Pond, Z.A., Burnett, R.T., 2019. Fine particulate air pollution
and human mortality: 25+ years of cohort studies. Environ. Res 108924.
Poujol-Robert, A., Boëlle, P.-Y., Conti, F., Durand, F., Duvoux, C., Wendum, D., Paradis,
V., Mackiewicz, V., Chazouillères, O., Corpechot, C., Poupon, R., 2014. Aspirin may
reduce liver brosis progression: Evidence from a multicenter retrospective study of
recurrent hepatitis C after liver transplantation. Clin. Res. Hepatol. Gastroenterol. 38,
570576.
Ptitsyn, A., Hulver, M., Cefalu, W., York, D., Smith, S.R., 2006. Unsupervised clustering of
gene expression data points at hypoxia as possible trigger for metabolic syndrome.
BMC Genomics 7, 318.
Puca, A.A., Carrizzo, A., Villa, F., Ferrario, A., Casaburo, M., Maciąg, A., Vecchione, C.,
2013. Vascular ageing: the role of oxidative stress. Int. J. Biochem. Cell Biol. 45,
556559.
Putin, E., Mamoshina, P., Aliper, A., Korzinkin, M., Moskalev, A., Kolosov, A., Ostrovskiy,
A., Cantor, C., Vijg, J., Zhavoronkov, A., 2016. Deep biomarkers of human aging:
Application of deep neural networks to biomarker development. Aging 8, 10211033.
Pyrkov, T.V., Slipensky, K., Barg, M., Kondrashin, A., Zhurov, B., Zenin, A., Pyatnitskiy,
M., Menshikov, L., Markov, S., Fedichev, P.O., 2018. Extracting biological age from
biomedical data via deep learning: too much of a good thing? Sci. Rep. 8, 5210.
Qiao, Y.-C., Chen, Y.-L., Pan, Y.-H., Ling, W., Tian, F., Zhang, X.-X., Zhao, H.-L., 2017.
Changes of transforming growth factor beta 1 in patients with type 2 diabetes and
diabetic nephropathy: A PRISMA-compliant systematic review and meta-analysis.
Medicine 96, e6583.
Qiu, W.Q., Zhu, H., 2014. Amylin and its analogs: a friend or foe for the treatment of
Alzheimers disease? Front. Aging Neurosci. 6, 186.
Quach, A., Levine, M.E., Tanaka, T., Lu, A.T., Chen, B.H., Ferrucci, L., Ritz, B., Bandinelli,
S., Neuhouser, M.L., Beasley, J.M., Snetselaar, L., Wallace, R.B., Tsao, P.S., Absher,
D., Assimes, T.L., Stewart, J.D., Li, Y., Hou, L., Baccarelli, A.A., Whitsel, E.A.,
Horvath, S., 2017. Epigenetic clock analysis of diet, exercise, education, and lifestyle
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
23
factors. Aging 9, 419446.
Rabbani, N., Adaikalakoteswari, A., Rossing, K., Rossing, P., Tarnow, L., Parving, H.-H.,
Thornalley, P.J., 2012. Eect of Irbesartan treatment on plasma and urinary markers
of protein damage in patients with type 2 diabetes and microalbuminuria. Amino
Acids 42, 16271639.
Radak, Z., Zhao, Z., Koltai, E., Ohno, H., Atalay, M., 2013. Oxygen Consumption and
Usage During Physical Exercise: The Balance Between Oxidative Stress and ROS-
Dependent Adaptive Signaling. Antioxid. Redox Signal. 18, 1208.
Rae, N., Golpour Hamedani, S., Barak, F., Safavi, S.M., Miraghajani, M., 2017. Dietary
patterns, food groups and telomere length: a systematic review of current studies.
Eur. J. Clin. Nutr. 71, 151158.
Rahman, S.A., Adjeroh, D.A., 2019. Deep Learning using Convolutional LSTM estimates
Biological Age from Physical Activity. Sci. Rep. 9, 11425.
Ramirez, C.E., Nian, H., Yu, C., Gamboa, J.L., Luther, J.M., Brown, N.J., Shibao, C.A.,
2015. Treatment with Sildenal Improves Insulin Sensitivity in Prediabetes: A
Randomized, Controlled Trial. J. Clin. Endocrinol. Metab. 100, 4533.
Ramirez-Sanchez, I., Taub, P.R., Ciaraldi, T.P., Nogueira, L., Coe, T., Perkins, G., Hogan,
M., Maisel, A.S., Henry, R.R., Ceballos, G., Villarreal, F., 2013. (-)-Epicatechin rich
cocoa mediated modulation of oxidative stress regulators in skeletal muscle of heart
failure and type 2 diabetes patients. Int. J. Cardiol. 168, 39823990.
Ravindrarajah, R., Lee, D.M., Pye, S.R., Gielen, E., Boonen, S., Vanderschueren, D.,
Pendleton, N., Finn, J.D., Tajar, A., OConnell, M.D.L., Rockwood, K., Bartfai, G.,
Casanueva, F.F., Forti, G., Giwercman, A., Han, T.S., Huhtaniemi, I.T., Kula, K., Lean,
M.E.J., Punab, M., Wu, F.C.W., ONeill, T.W., European Male Aging Study Group,
2013. The ability of three dierent models of frailty to predict all-cause mortality:
results from the European Male Aging Study (EMAS). Arch. Gerontol. Geriatr. 57,
360368.
Raygan, F., Ostadmohammadi, V., Bahmani, F., Reiter, R.J., Asemi, Z., 2017. Melatonin
administration lowers biomarkers of oxidative stress and cardio-metabolic risk in
type 2 diabetic patients with coronary heart disease: A randomized, double-blind,
placebo-controlled trial. Clin. Nutr. https://doi.org/10.1016/j.clnu.2017.12.004.
Raymond, I., 2003. The inuence of age, sex and other variables on the plasma level of N-
terminal pro brain natriuretic peptide in a large sample of the general population.
Heart. https://doi.org/10.1136/heart.89.7.745.
Reddy, S.P., Handa, A., Tan, L., Devaney, A., Hughes, D., Mason, P., Friend, P.J., Darby,
C.R., 2003. Low-dose valaciclovir prophylaxis against cytomegalovirus disease in
renal transplant recipients. Transpl. Int. 16, 726729.
Regazzoni, L., de Courten, B., Garzon, D., Altomare, A., Marinello, C., Jakubova, M.,
Vallova, S., Krumpolec, P., Carini, M., Ukropec, J., Ukropcova, B., Aldini, G., 2016. A
carnosine intervention study in overweight human volunteers: bioavailability and
reactive carbonyl species sequestering eect. Sci. Rep. 6, 27224.
Reichert, S., Stier, A., 2017. Does oxidative stress shorten telomeres in vivo? A review.
Biol. Lett. 13. https://doi.org/10.1098/rsbl.2017.0463.
Reinisalo, M., Kårlund, A., Koskela, A., Kaarniranta, K., Karjalainen, R.O., 2015.
Polyphenol Stilbenes: Molecular Mechanisms of Defence against Oxidative Stress and
Aging-Related Diseases. Oxid. Med. Cell. Longev., 340520 2015.
Revelas, M., Thalamuthu, A., Oldmeadow, C., Evans, T.-J., Armstrong, N.J., Kwok, J.B.,
Brodaty, H., Schoeld, P.R., Scott, R.J., Sachdev, P.S., Attia, J.R., Mather, K.A., 2018.
Review and meta-analysis of genetic polymorphisms associated with exceptional
human longevity. Mech. Ageing Dev. 175, 2434.
Rhee, E.-J., Kim, H.C., Kim, J.H., Lee, E.Y., Kim, B.J., Kim, E.M., Song, Y., Lim, J.H., Kim,
H.J., Choi, S., Moon, M.K., Na, J.O., Park, K.-Y., Oh, M.S., Han, S.Y., Noh, J., Yi, K.H.,
Lee, S.-H., Hong, S.-C., Jeong, I.-K., 2019. 2018 Guidelines for the management of
dyslipidemia. Korean J. Intern. Med. 34, 723771.
Richman, I.B., Owens, D.K., 2017. Aspirin for Primary Prevention. Medical Clinics 101,
713724.
Rico-Uribe, L.A., Caballero, F.F., Martín-María, N., Cabello, M., Ayuso-Mateos, J.L., Miret,
M., 2018. Association of loneliness with all-cause mortality: A meta-analysis. PLoS
One 13, e0190033.
Ridker, P.M., Lüscher, T.F., 2014. Anti-inammatory therapies for cardiovascular disease.
Eur. Heart J. 35, 17821791.
Riekse, R.G., Li, G., Petrie, E.C., Leverenz, J.B., Vavrek, D., Vuletic, S., Albers, J.J.,
Montine, T.J., Lee, V.M.-Y., Lee, M., Seubert, P., Galasko, D., Schellenberg, G.D.,
Hazzard, W.R., Peskind, E.R., 2006. Eect of statins on Alzheimers disease bio-
markers in cerebrospinal uid. J. Alzheimers. Dis. 10, 399406.
Rienks, J., Barbaresko, J., Nöthlings, U., 2017. Association of Polyphenol Biomarkers with
Cardiovascular Disease and Mortality Risk: A Systematic Review and Meta-Analysis
of Observational Studies. Nutrients 9. https://doi.org/10.3390/nu9040415.
Rockwood, K., Mitnitski, A., 2007. Frailty in relation to the accumulation of decits. J.
Gerontol. A Biol. Sci. Med. Sci. 62, 722727.
Rockwood, K., Song, X., MacKnight, C., Bergman, H., Hogan, D.B., McDowell, I.,
Mitnitski, A., 2005. A global clinical measure of tness and frailty in elderly people.
CMAJ 173, 489495.
Rockwood, K., Song, X., Mitnitski, A., 2011. Changes in relative tness and frailty across
the adult lifespan: evidence from the Canadian National Population Health Survey.
CMAJ 183, E487.
Rodrigue, K.M., Kennedy, K.M., Park, D.C., 2009. Beta-amyloid deposition and the aging
brain. Neuropsychol. Rev. 19, 436450.
Rogers, N.T., Demakakos, P., Taylor, M.S., Steptoe, A., Hamer, M., Shankar, A., 2016.
Volunteering is associated with increased survival in able-bodied participants of the
English Longitudinal Study of Ageing. J. Epidemiol. Community Health 70, 583.
Rogina, B., 2017. INDYA New Link to Metabolic Regulation in Animals and Humans.
Front. Genet. 8. https://doi.org/10.3389/fgene.2017.00066.
Rojas, J., Chávez-Castillo, M., Olivar, L.C., Calvo, M., Mejías, J., Rojas, M., Morillo, J.,
Bermúdez, V., 2015. Physiologic Course of Female Reproductive Function: A
Molecular Look into the Prologue of Life. J. Pregnancy, 715735 2015.
Romero-Ortuno, R., Kenny, R.A., 2012. The frailty index in Europeans: association with
age and mortality. Age Ageing 41, 5.
Rosano, A., Dai, Q., Shapses, S.A., 2016. Essential Nutrient Interactions: Does Low or
Suboptimal Magnesium Status Interact with Vitamin D and/or Calcium Status? Adv.
Nutr. 7, 25.
Rosenblit, P.D., 2016. Common medications used by patients with type 2 diabetes mel-
litus: what are their eects on the lipid prole? Cardiovasc. Diabetol. 15. https://doi.
org/10.1186/s12933-016-0412-7.
Rosenbloom, J., Macarak, E., Piera-Velazquez, S., Jimenez, S.A., 2017. Human Fibrotic
Diseases: Current Challenges in Fibrosis Research. Methods Mol. Biol. 1627, 123.
Rosero-Bixby, L., Dow, W.H., 2012. Predicting mortality with biomarkers: a population-
based prospective cohort study for elderly Costa Ricans. Popul. Health Metr. 10, 11.
Rossman, M.J., Santos-Parker, J.R., Steward, C.A.C., Bispham, N.Z., Cuevas, L.M.,
Rosenberg, H.L., Woodward, K.A., Chonchol, M., Gioscia-Ryan, R.A., Murphy, M.P.,
Seals, D.R., 2018. Chronic Supplementation With a Mitochondrial Antioxidant
(MitoQ) Improves Vascular Function in Healthy Older Adults. Hypertension 71,
10561063.
Roy, S., Amin, S., Roy, S., 2016. Retinal brosis in diabetic retinopathy. Exp. Eye Res.
142, 7175.
Ruegsegger, G.N., Booth, F.W., 2017. Running from Disease: Molecular Mechanisms
Associating Dopamine and Leptin Signaling in the Brain with Physical Inactivity,
Obesity, and Type 2 Diabetes. Front. Endocrinol. 8. https://doi.org/10.3389/fendo.
2017.00109.
Sae-Lee, C., Corsi, S., Barrow, T.M., Kuhnle, G.G.C., Bollati, V., Mathers, J.C., Byun, H.-
M., 2018. Dietary Intervention Modies DNA Methylation Age Assessed by the
Epigenetic Clock. Mol. Nutr. Food Res. 62, e1800092.
Sahebkar, A., 2017. Eects of quercetin supplementation on lipid prole: A systematic
review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr.
57, 666676.
Sahebkar, A., Simental-Mendía, L.E., Reiner, Ž., Kovanen, P.T., Simental-Mendía, M.,
Bianconi, V., Pirro, M., 2017. Eect of orlistat on plasma lipids and body weight: A
systematic review and meta-analysis of 33 randomized controlled trials. Pharmacol.
Res. 122, 5365.
Salas-Pérez, F., Ramos-Lopez, O., Mansego, M.L., Milagro, F.I., Santos, J.L., Riezu-Boj,
J.I., Martínez, J.A., 2019. DNA methylation in genes of longevity-regulating path-
ways: association with obesity and metabolic complications. Aging 11, 18741899.
Samani, N.B., Jokar, A., Soveid, M., Heydari, M., Mosavat, S.H., 2016. Ecacy of the
Hydroalcoholic Extract of Tribulus terrestris on the Serum Glucose and Lipid Prole
of Women With Diabetes Mellitus: A Double-Blind Randomized Placebo-Controlled
Clinical Trial. J. Evid. Based Complementary Altern. Med. 21 NP917.
Santi, D., Giannetta, E., Isidori, A.M., Vitale, C., Aversa, A., Simoni, M., 2015. Therapy of
endocrine disease. Eects of chronic use of phosphodiesterase inhibitors on en-
dothelial markers in type 2 diabetes mellitus: a meta-analysis. Eur. J. Endocrinol.
172, R10314.
Sapre, S., Thakur, R., 2014. Lifestyle and dietary factors determine age at natural me-
nopause. J. Midlife. Health 5, 35.
Saxton, R.A., Sabatini, D.M., 2017. mTOR Signaling in Growth, Metabolism, and Disease.
Cell 169, 361371.
Schoenaker, D.A.J.M., Jackson, C.A., Rowlands, J.V., Mishra, G.D., 2014. Socioeconomic
position, lifestyle factors and age at natural menopause: a systematic review and
meta-analyses of studies across six continents. Int. J. Epidemiol. 43, 15421562.
Schuliga, M., Pechkovsky, D.V., Read, J., Waters, D.W., Blokland, K.E.C., Reid, A.T.,
Hogaboam, C.M., Khalil, N., Burgess, J.K., Prêle, C.M., Mutsaers, S.E., Jaar, J.,
Westall, G., Grainge, C., Knight, D.A., 2018. Mitochondrial dysfunction contributes to
the senescent phenotype of IPF lung broblasts. J. Cell. Mol. Med. 22, 58475861.
Schur, E.A., Melhorn, S.J., Oh, S.-K., Lacy, J.M., Berkseth, K.E., Guyenet, S.J., Sonnen,
J.A., Tyagi, V., Rosalynn, M., De Leon, B., Webb, M.F., Gonsalves, Z.T., Fligner, C.L.,
Schwartz, M.W., Maravilla, K.R., 2015. Radiologic evidence that hypothalamic gliosis
is associated with obesity and insulin resistance in humans. Obesity 23, 21422148.
Schwartz, A.V., 2015. Marrow fat and bone: review of clinical ndings. Front. Endocrinol.
6, 40.
Schwingshackl, L., Chaimani, A., Homann, G., Schwedhelm, C., Boeing, H., 2018a. A
network meta-analysis on the comparative ecacy of dierent dietary approaches on
glycaemic control in patients with type 2 diabetes mellitus. Eur. J. Epidemiol. 33,
157.
Schwingshackl, L., Christoph, M., Homann, G., 2015. Eects of Olive Oil on Markers of
Inammation and Endothelial Function-A Systematic Review and Meta-Analysis.
Nutrients 7, 76517675.
Schwingshackl, L., Homann, G., 2014. Monounsaturated fatty acids, olive oil and health
status: a systematic review and meta-analysis of cohort studies. Lipids Health Dis. 13,
154.
Schwingshackl, L., Homann, G., Iqbal, K., Schwedhelm, C., Boeing, H., 2018b. Food
groups and intermediate disease markers: a systematic review and network meta-
analysis of randomized trials. Am. J. Clin. Nutr. 108, 576586.
Schwingshackl, L., Homann, G., Lampousi, A.-M., Knüppel, S., Iqbal, K., Schwedhelm,
C., Bechthold, A., Schlesinger, S., Boeing, H., 2017a. Food groups and risk of type 2
diabetes mellitus: a systematic review and meta-analysis of prospective studies. Eur.
J. Epidemiol. 32, 363.
Schwingshackl, L., Lampousi, A.-M., Portillo, M.P., Romaguera, D., Homann, G., Boeing,
H., 2017b. Olive oil in the prevention and management of type 2 diabetes mellitus: a
systematic review and meta-analysis of cohort studies and intervention trials. Nutr.
Diabetes 7, e262.
Schwingshackl, L., Schwedhelm, C., Homann, G., Lampousi, A.-M., Knüppel, S., Iqbal,
K., Bechthold, A., Schlesinger, S., Boeing, H., 2017c. Food groups and risk of all-cause
mortality: a systematic review and meta-analysis of prospective studies. Am. J. Clin.
Nutr. 105, 14621473.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
24
Sebastiani, P., Thyagarajan, B., Sun, F., Honig, L.S., Schupf, N., Cosentino, S., Feitosa,
M.F., Wojczynski, M., Newman, A.B., Montano, M., Perls, T.T., 2016. Age and Sex
Distributions of Age-Related Biomarker Values in Healthy Older Adults from the Long
Life Family Study. J. Am. Geriatr. Soc. 64, e189e194.
Sebastiani, P., Thyagarajan, B., Sun, F., Schupf, N., Newman, A.B., Montano, M., Perls,
T.T., 2017. Biomarker signatures of aging. Aging Cell 16, 329338.
Seely, D., Wu, P., Fritz, H., Kennedy, D.A., Tsui, T., Seely, A.J.E., Mills, E., 2012.
Melatonin as adjuvant cancer care with and without chemotherapy: a systematic
review and meta-analysis of randomized trials. Integr. Cancer Ther. 11, 293303.
Serban, M., Sahebkar, A., Zanchetti, A., Mikhailidis, D.P., Howard, G., Antal, D., Andrica,
F., Ahmed, A., Aronow, W.S., Muntner, P., Lip, G.Y.H., Graham, I., Wong, N., Rysz, J.,
Banach, M., Lipid, F.T., Blood Pressure Metaanalysis Collaboration (LBPMC) Group,
2016. Eects of Quercetin on Blood Pressure: A Systematic Review and MetaAnalysis
of Randomized Controlled Trials. Journal of the American Heart Association:
Cardiovascular and Cerebrovascular Disease 5. https://doi.org/10.1161/JAHA.115.
002713.
Shan, W., Liu, J., 2009. Inammation: a hidden path to breaking the spell of ovarian
cancer. Cell Cycle 8, 31073111.
Shapiro, B.B., Streja, E., Chen, J.L.T., Kovesdy, C.P., Kalantar-Zadeh, K., Rhee, C.M.,
2014. The relationship between ultraviolet light exposure and mortality in dialysis
patients. Am. J. Nephrol. 40, 224232.
Shen, H., Shahzad, G., Jawairia, M., Bostick, R.M., Mustacchia, P., 2014. Association
between aspirin use and the prevalence of nonalcoholic fatty liver disease: a cross-
sectional study from the Third National Health and Nutrition Examination Survey.
Aliment. Pharmacol. Ther. 40, 10661073.
Shi, F., Kouadir, M., Yang, Y., 2015. NALP3 inammasome activation in protein mis-
folding diseases. Life Sci. 135, 914.
Shigiyama, F., Kumashiro, N., Miyagi, M., Ikehara, K., Kanda, E., Uchino, H., Hirose, T.,
2017. Eectiveness of dapagliozin on vascular endothelial function and glycemic
control in patients with early-stage type 2 diabetes mellitus: DEFENCE study.
Cardiovasc. Diabetol. 16. https://doi.org/10.1186/s12933-017-0564-0.
Shores, M.M., Matsumoto, A.M., 2014. Testosterone, aging and survival. Curr. Opin.
Endocrinol. Diabetes Obes. 21, 209216.
Short, K.R., Vittone, J.L., Bigelow, M.L., Proctor, D.N., Nair, K.S., 2004. Age and aerobic
exercise training eects on whole body and muscle protein metabolism. Short, Kevin
R; Vittone, Janet L; Bigelow, Maureen L; Proctor, David N; Nair, K Sreekumaran 286.
pp. E92101.
Shrikrishna, D., Astin, R., Kemp, P.R., Hopkinson, N.S., 2012. Renin-angiotensin system
blockade: a novel therapeutic approach in chronic obstructive pulmonary disease.
Clin. Sci. 123, 487498.
Shulman, G.I., 2014. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic
disease. N. Engl. J. Med. 371, 11311141.
Silvestre, M.P., Goode, J.P., Vlaskovsky, P., McMahon, C., Tay, A., Poppitt, S.D., 2018.
The role of glucagon in weight loss-mediated metabolic improvement: a systematic
review and meta-analysis. Obes. Rev. 19, 233253.
Simental-Mendia, L.E., Sahebkar, A., Rodriguez-Moran, M., Zambrano-Galvan, G.,
Guerrero-Romero, F., 2017. Eect of Magnesium Supplementation on Plasma C-re-
active Protein Concentrations: A Systematic Review and Meta-Analysis of
Randomized Controlled Trials. Curr. Pharm. Des. 23, 46784686.
Simpson, R.J., Cosgrove, C., Chee, M.M., McFarlin, B.K., Bartlett, D.B., Spielmann, G.,
OConnor, D.P., Pircher, H., Shiels, P.G., 2010. Senescent phenotypes and telomere
lengths of peripheral blood T-cells mobilized by acute exercise in humans. Exerc.
Immunol. Rev. 16, 4055.
Singh, S., Trikha, S., Bhowmick, D.C., Sarkar, A.A., Jeremic, A.M., 2015. Role of
Cholesterol and Phospholipids in Amylin Misfolding, Aggregation and Etiology of
Islet Amyloidosis. Adv. Exp. Med. Biol. 855, 95.
Siparsky, P.N., Kirkendall, D.T., Garrett Jr, W.E., 2014. Muscle changes in aging: un-
derstanding sarcopenia. Sports Health 6, 3640.
Smeuninx, B., Mckendry, J., Wilson, D., Martin, U., Breen, L., 2017. Age-Related Anabolic
Resistance of Myobrillar Protein Synthesis Is Exacerbated in Obese Inactive
Individuals. J. Clin. Endocrinol. Metab. 102, 35353545.
Smith, E.R., Tomlinson, L.A., Ford, M.L., McMahon, L.P., Rajkumar, C., Holt, S.G., 2012.
Elastin degradation is associated with progressive aortic stiening and all-cause
mortality in predialysis chronic kidney disease. Hypertension 59, 973978.
Smith, K.B., Kang, P., Sabbagh, M.N., Disease Neuroimaging Initiative, Alzheimers, 2017.
The Eect of Statins on Rate of Cognitive Decline in Mild Cognitive Impairment.
Alzheimers. Dement. 3, 149156.
Smith, W.C., Anderson, E., Salinas, D., Horvatek, R., Baker, D.P., 2015. A meta-analysis of
education eects on chronic disease: the causal dynamics of the Population Education
Transition Curve. Soc. Sci. Med. 127, 2940.
Snyder, P.J., Bhasin, S., Cunningham, G.R., Matsumoto, A.M., Stephens-Shields, A.J.,
Cauley, J.A., Gill, T.M., Barrett-Connor, E., Swerdlo, R.S., Wang, C., Ensrud, K.E.,
Lewis, C.E., Farrar, J.T., Cella, D., Rosen, R.C., Pahor, M., Crandall, J.P., Molitch,
M.E., Cifelli, D., Dougar, D., Fluharty, L., Resnick, S.M., Storer, T.W., Anton, S.,
Basaria, S., Diem, S.J., Hou, X., Mohler 3rd, E.R., Parsons, J.K., Wenger, N.K., Zeldow,
B., Landis, J.R., Ellenberg, S.S., Testosterone Trials Investigators, 2016. Eects of
Testosterone Treatment in Older Men. N. Engl. J. Med. 374, 611624.
Soltani, S., Chitsazi, M.J., Salehi-Abargouei, A., 2017. The eect of dietary approaches to
stop hypertension (DASH) on serum inammatory markers: A systematic review and
meta-analysis of randomized trials. Clin. Nutr. https://doi.org/10.1016/j.clnu.2017.
02.018.
Sørensen, C.E., Tritsaris, K., Reibel, J., Lauritzen, M., Mortensen, E.L., Osler, M., Pedersen,
A.M.L., 2016. Elevated p16ink4a Expression in Human Labial Salivary Glands as a
Potential Correlate of Cognitive Aging in Late Midlife. PLoS One 11, e0152612.
Soysal, P., Stubbs, B., Lucato, P., Luchini, C., Solmi, M., Peluso, R., Sergi, G., Isik, A.T.,
Manzato, E., Maggi, S., Maggio, M., Prina, A.M., Cosco, T.D., Wu, Y.-T., Veronese, N.,
2016. Inammation and frailty in the elderly: A systematic review and meta-analysis.
Ageing Res. Rev. 31, 18.
Spendi, S., Vuda, M., Gouspillou, G., Aare, S., Perez, A., Morais, J.A., Jagoe, R.T., Filion,
M.-E., Glicksman, R., Kapchinsky, S., MacMillan, N.J., Pion, C.H., Aubertin-Leheudre,
M., Hettwer, S., Correa, J.A., Taivassalo, T., Hepple, R.T., 2016. Denervation drives
mitochondrial dysfunction in skeletal muscle of octogenarians. J. Physiol. 594,
73617379.
Stambler, I., 2017. Recognizing Degenerative Aging as a Treatable Medical Condition:
Methodology and Policy. Aging Dis. 8, 583589.
Stoner, L., Rowlands, D., Morrison, A., Credeur, D., Hamlin, M., Ganey, K., Lambrick, D.,
Matheson, A., 2016. Ecacy of Exercise Intervention for Weight Loss in Overweight
and Obese Adolescents: Meta-Analysis and Implications. Sports Med. 46, 17371751.
Strazhesko, I.D., Tkacheva, O.N., Akasheva, D.U., Dudinskaya, E.N., Plokhova, E.V.,
Pykhtina, V.S., Kruglikova, A.S., Kokshagina, N.V., Sharashkina, N.V., Agaltsov, M.V.,
Kashtanova, D.A., Vygodin, V.A., Ozerova, I.N., Skvortsov, D.A., Vasilkova, D.,
Boytsov, S.A., 2016. Atorvastatin Therapy Modulates Telomerase Activity in Patients
Free of Atherosclerotic Cardiovascular Diseases. Front. Pharmacol. 7. https://doi.
org/10.3389/fphar.2016.00347.
Strazhesko, I., Tkacheva, O., Boytsov, S., Akasheva, D., Dudinskaya, E., Vygodin, V.,
Skvortsov, D., Nilsson, P., 2015. Association of Insulin Resistance, Arterial Stiness
and Telomere Length in Adults Free of Cardiovascular Diseases. PLoS One 10,
e0136676.
Stringhini, S., Carmeli, C., Jokela, M., Avendaño, M., Muennig, P., Guida, F., Ricceri, F.,
dErrico, A., Barros, H., Bochud, M., Chadeau-Hyam, M., Clavel-Chapelon, F., Costa,
G., Delpierre, C., Fraga, S., Goldberg, M., Giles, G.G., Krogh, V., Kelly-Irving, M.,
Layte, R., Lasserre, A.M., Marmot, M.G., Preisig, M., Shipley, M.J., Vollenweider, P.,
Zins, M., Kawachi, I., Steptoe, A., Mackenbach, J.P., Vineis, P., Kivimäki, M.,
LIFEPATH consortium, 2017. Socioeconomic status and the 25 × 25 risk factors as
determinants of premature mortality: a multicohort study and meta-analysis of 1·7
million men and women. Lancet 389, 12291237.
Sui, B., Hu, C., Jin, Y., 2016. Mitochondrial metabolic failure in telomere attrition-pro-
voked aging of bone marrow mesenchymal stem cells. Biogerontology 17, 267279.
Su, J., Ekman, C., Oskolkov, N., Lahti, L., Ström, K., Brazma, A., Groop, L., Rung, J.,
Hansson, O., 2015. A novel atlas of gene expression in human skeletal muscle reveals
molecular changes associated with aging. Skelet. Muscle 5, 35.
Suliman, H.B., Piantadosi, C.A., 2016. Mitochondrial Quality Control as a Therapeutic
Target. Pharmacol. Rev. 68, 2048.
Sutclie, P., Connock, M., Gurung, T., Freeman, K., Johnson, S., Kandala, N.-B., Grove, A.,
Gurung, B., Morrow, S., Clarke, A., 2013. Aspirin for prophylactic use in the primary
prevention of cardiovascular disease and cancer: a systematic review and overview of
reviews. Health Technol. Assess. 17, 1253.
Suzuki, S., Nishio, S.-I., Takeda, T., Komatsu, M., 2012. Gender-specic regulation of
response to thyroid hormone in aging. Thyroid Res. 5, 1.
Szcześniak, D., Budzeń, S., Kopeć, W., Rymaszewska, J., 2014. Anserine and carnosine
supplementation in the elderly: Eects on cognitive functioning and physical capa-
city. Arch. Gerontol. Geriatr. 59, 485490.
Szekely, A., Kotyuk, E., Bircher, J., Vereczkei, A., Balota, D.A., Sasvari-Szekely, M., Ronai,
Z., 2016. Association between Age and the 7 Repeat Allele of the Dopamine D4
Receptor Gene. PLoS One 11, e0167753.
Tabrizi, R., Tamtaji, O.R., Lankarani, K.B., Mirhosseini, N., Akbari, M., Dadgostar, E.,
Peymani, P., Asemi, Z., 2018a. The eects of resveratrol supplementation on bio-
markers of inammation and oxidative stress among patients with metabolic syn-
drome and related disorders: a systematic review and meta-analysis of randomized
controlled trials. Food Funct. 9, 61166128.
Tabrizi, R., Vakili, S., Akbari, M., Mirhosseini, N., Lankarani, K.B., Rahimi, M., Mobini,
M., Jafarnejad, S., Vahedpoor, Z., Asemi, Z., 2018b. The eects of curcumin-con-
taining supplements on biomarkers of inammation and oxidative stress: A sys-
tematic review and meta-analysis of randomized controlled trials. Phytother. Res.
https://doi.org/10.1002/ptr.6226.
Tajar, A., OConnell, M.D.L., Mitnitski, A.B., ONeill, T.W., Searle, S.D., Huhtaniemi, I.T.,
Finn, J.D., Bartfai, G., Boonen, S., Casanueva, F.F., Forti, G., Giwercman, A., Han,
T.S., Kula, K., Labrie, F., Lean, M.E.J., Pendleton, N., Punab, M., Silman, A.J.,
Vanderschueren, D., Rockwood, K., Wu, F.C.W., European Male Aging Study Group,
2011. Frailty in relation to variations in hormone levels of the hypothalamic-pitui-
tary-testicular axis in older men: results from the European male aging study. J. Am.
Geriatr. Soc. 59, 814821.
Takagi, H., Mizuno, Y., Yamamoto, H., Goto, S.-N., Umemoto, T., All-Literature
Investigation of Cardiovascular Evidence Group, 2013. Eects of telmisartan therapy
on interleukin-6 and tumor necrosis factor-alpha levels: a meta-analysis of rando-
mized controlled trials. Hypertens. Res. 36, 368373.
Takeuchi, M., Takino, J.-I., Sakasai-Sakai, A., Takata, T., Tsutsumi, M., 2017. Toxic AGE
(TAGE) Theory for the Pathophysiology of the Onset/Progression of NAFLD and ALD.
Nutrients 9. https://doi.org/10.3390/nu9060634.
Talasaz, A.H., Khalili, H., Fahimi, F., Jenab, Y., Broumand, M.A., Salarifar, M., Darabi, F.,
2014. Eects of N-acetylcysteine on the cardiac remodeling biomarkers and major
adverse events following acute myocardial infarction: a randomized clinical trial.
Am. J. Cardiovasc. Drugs 14, 5161.
Tardito, E., 1990. Vascular abnormalities of the limbs. Some observations on the classi-
cation. Minerva Chir. 45, 953955.
Taylor, E.L., Armstrong, K.R., Perrett, D., Hattersley, A.T., Winyard, P.G., 2015.
Optimisation of an Advanced Oxidation Protein Products Assay: Its Application to
Studies of Oxidative Stress in Diabetes Mellitus. Oxid. Med. Cell. Longev. https://doi.
org/10.1155/2015/496271. 2015.
Tian, H., Lu, J., He, H., Zhang, L., Dong, Y., Yao, H., Feng, W., Wang, S., 2016. The eect
of Astragalus as an adjuvant treatment in type 2 diabetes mellitus: A (preliminary)
meta-analysis. J. Ethnopharmacol. 191, 206215.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
25
Toth, P., Tarantini, S., Csiszar, A., Ungvari, Z., 2017. Functional vascular contributions to
cognitive impairment and dementia: mechanisms and consequences of cerebral au-
toregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in
aging. Am. J. Physiol. Heart Circ. Physiol. 312, H1H20.
Trammell, S.A.J., Schmidt, M.S., Weidemann, B.J., Redpath, P., Jaksch, F., Dellinger,
R.W., Li, Z., Abel, E.D., Migaud, M.E., Brenner, C., 2016. Nicotinamide riboside is
uniquely and orally bioavailable in mice and humans. Nat. Commun. 7, 12948.
Uchoa, M.F., Moser, V.A., Pike, C.J., 2016. Interactions between inammation, sex
steroids, and Alzheimers disease risk factors. Front. Neuroendocrinol. 43, 6082.
Ungvari, Z., Tarantini, S., Donato, A.J., Galvan, V., Csiszar, A., 2018. Mechanisms of
Vascular Aging. Circ. Res. 123, 849867.
Uribarri, J., Peppa, M., Cai, W., Goldberg, T., Lu, M., He, C., Vlassara, H., 2003.
Restriction of dietary glycotoxins reduces excessive advanced glycation end products
in renal failure patients. J. Am. Soc. Nephrol. 14, 728731.
Van Acker, H.H., Anguille, S., Willemen, Y., Smits, E.L., Van Tendeloo, V.F., 2016.
Bisphosphonates for cancer treatment: Mechanisms of action and lessons from clin-
ical trials. Pharmacol. Ther. 158, 2440.
van de Weijer, T., Phielix, E., Bilet, L., Williams, E.G., Ropelle, E.R., Bierwagen, A.,
Livingstone, R., Nowotny, P., Sparks, L.M., Paglialunga, S., Szendroedi, J., Havekes,
B., Moullan, N., Pirinen, E., Hwang, J.-H., Schrauwen-Hinderling, V.B., Hesselink,
M.K.C., Auwerx, J., Roden, M., Schrauwen, P., 2015. Evidence for a direct eect of
the NAD+ precursor acipimox on muscle mitochondrial function in humans.
Diabetes 64, 11931201.
Vanhooren, V., Dewaele, S., Libert, C., Engelborghs, S., De Deyn, P.P., Toussaint, O.,
Debacq-Chainiaux, F., Poulain, M., Glupczynski, Y., Franceschi, C., Jaspers, K., van
der Pluijm, I., Hoeijmakers, J., Chen, C.C., 2010. Serum N-glycan prole shift during
human ageing. Exp. Gerontol. 45, 738743.
Vega, R.B., Horton, J.L., Kelly, D.P., 2015. Maintaining ancient organelles: mitochondrial
biogenesis and maturation. Circ. Res. 116, 18201834.
Veldhuis, J.D., Keenan, D.M., Liu, P.Y., Iranmanesh, A., Takahashi, P.Y., Nehra, A.X.,
2009. The aging male hypothalamicpituitarygonadal axis: Pulsatility and feedback.
Mol. Cell. Endocrinol. 299, 1422.
Velissaris, D., Pantzaris, N., Koniari, I., Koutsogiannis, N., Karamouzos, V., Kotroni, I.,
Skroumpelou, A., Ellul, J., 2017. C-Reactive Protein and Frailty in the Elderly: A
Literature Review. J. Clin. Med. Res. 9, 461465.
Verma, H., Garg, R., 2017. Eect of magnesium supplementation on type 2 diabetes as-
sociated cardiovascular risk factors: a systematic review and meta-analysis. J. Hum.
Nutr. Diet. 30, 621633.
Verrijken, A., Francque, S., Van Gaal, L., Science, M. of, Department of Endocrinology,
Metabolism, D.A, Hepatologist, Department of Gastroenterology and Hepatology,
Head, of M.A, Department of Endocrinology, Metabolism, D.A, Antwerp University
Hospital, University of Antwerp, 2010. The Role of Visceral Adipose Tissue in the
Pathogenesis of Non-alcoholic Fatty Liver Disease. European Endocrinology 7, 96.
Vescovini, R., Fagnoni, F.F., Telera, A.R., Bucci, L., Pedrazzoni, M., Magalini, F., Stella, A.,
Pasin, F., Medici, M.C., Calderaro, A., Volpi, R., Monti, D., Franceschi, C., Nikolich-
Žugich, J., Sansoni, P., 2014. Naïve and memory CD8 T cell pool homeostasis in
advanced aging: impact of age and of antigen-specic responses to cytomegalovirus.
Age 36, 625640.
Vestergaard, Esben, et al., 2017. Short-term acipimox treatment is associated with de-
creased cardiac parasympathetic modulation. Br. J. Clin. Pharmacol. 83 (12),
26712677. https://doi.org/10.1111/bcp.13384.
Viña, J., Borrás, C., Gambini, J., Sastre, J., Pallardó, F.V., 2005. Why females live longer
than males? Importance of the upregulation of longevity-associated genes by oes-
trogenic compounds. FEBS Lett. 579, 25412545.
Volkow, N.D., Tomasi, D., Wang, G.-J., Telang, F., Fowler, J.S., Goldstein, R.Z., Klein, N.,
Wong, C., Swanson, J.M., Shumay, E., 2013. Association between dopamine D4 re-
ceptor polymorphism and age related changes in brain glucose metabolism. PLoS One
8, e63492.
Wagner, K.-H., Cameron-Smith, D., Wessner, B., Franzke, B., 2016. Biomarkers of Aging:
From Function to Molecular Biology. Nutrients 8. https://doi.org/10.3390/
nu8060338.
Wang, J.C., Bennett, M., 2012. Aging and atherosclerosis: mechanisms, functional con-
sequences, and potential therapeutics for cellular senescence. Circ. Res. 111,
245259.
Wang, L., Mascher, H., Psilander, N., Blomstrand, E., Sahlin, K., 2011. Resistance exercise
enhances the molecular signaling of mitochondrial biogenesis induced by endurance
exercise in human skeletal muscle. J. Appl. Physiol. 111, 13351344.
Wang, M., Zhao, D., Spinetti, G., Zhang, J., Jiang, L.-Q., Pintus, G., Monticone, R.,
Lakatta, E.G., 2006. Matrix metalloproteinase 2 activation of transforming growth
factor-beta1 (TGF-beta1) and TGF-beta1-type II receptor signaling within the aged
arterial wall. Arterioscler. Thromb. Vasc. Biol. 26, 15031509.
Wang, Y., Chen, F., Ye, L., Zirkin, B., Chen, H., 2017. Steroidogenesis in Leydig cells:
eects of aging and environmental factors. Reproduction 154, R111R122.
Weakley, S.M., Jiang, J., Kougias, P., Lin, P.H., Yao, Q., Charles Brunicardi, F., Gibbs,
R.A., Chen, C., 2010. Role of Somatic Mutations in Vascular Disease Formation.
Expert Rev. Mol. Diagn. 10, 173.
Weidner, C.I., Lin, Q., Koch, C.M., Eisele, L., Beier, F., Ziegler, P., Bauerschlag, D.O.,
Jöckel, K.-H., Erbel, R., Mühleisen, T.W., Zenke, M., Brümmendorf, T.H., Wagner, W.,
2014. Aging of blood can be tracked by DNA methylation changes at just three CpG
sites. Genome Biol. 15, R24.
Weisberg, S.P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R.L., Ferrante, A.W., 2003.
Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest.
112, 17961808.
Weltevrede, M., Eilers, R., de Melker, H.E., van Baarle, D., 2016. Cytomegalovirus per-
sistence and T-cell immunosenescence in people aged fty and older: A systematic
review. Exp. Gerontol. 77, 8795.
Welty, F.K., 2013. How Do Elevated Triglycerides and Low HDL-Cholesterol Aect
Inammation and Atherothrombosis? Curr. Cardiol. Rep. 15, 400.
Wen, W.X., Lee, S.Y., Siang, R., Koh, R.Y., 2017. Repurposing Pentoxifylline for the
Treatment of Fibrosis: An Overview. Adv. Ther. 34, 12451269.
White, R.R., Vijg, J., 2016. Do DNA double-strand breaks drive aging? Mol. Cell 63, 729.
Whitlock, E.P., Williams, S.B., Burda, B.U., Feightner, A., Beil, T., 2015. Aspirin Use in
Adults: Cancer, All-Cause Mortality, and Harms: A Systematic Evidence Review for
the U.S. Preventive Services Task Force. Agency for Healthcare Research and Quality
(US), Rockville (MD).
Wiciński, M., Żak, J., Malinowski, B., Popek, G., Grześk, G., 2017. PCSK9 signaling
pathways and their potential importance in clinical practice. EPMA J. 8, 391.
Wikby, A., Maxson, P., Olsson, J., Johansson, B., Ferguson, F.G., 1998. Changes in CD8
and CD4 lymphocyte subsets, T cell proliferation responses and non-survival in the
very old: the Swedish longitudinal OCTO-immune study. Mech. Ageing Dev. 102,
187198.
Wilding, M., 2015. Potential long-term risks associated with maternal aging (the role of
the mitochondria). Fertil. Steril. 103, 13971401.
Wilkosz, P., Greggains, G.D., Tanbo, T.G., Fedorcsak, P., 2014. Female reproductive de-
cline is determined by remaining ovarian reserve and age. PLoS One 9, e108343.
Williams, B.R., Cho, J.S., 2017. Hormone Replacement: The Fountain of Youth? Prim.
Care 44, 481498.
Williams, S.L., Mash, D.C., Züchner, S., Moraes, C.T., 2013. Somatic mtDNA mutation
spectra in the aging human putamen. PLoS Genet. 9, e1003990.
Woods, D.C., Tilly, J.L., 2015. Autologous Germline Mitochondrial Energy Transfer
(AUGMENT) in Human Assisted Reproduction. Semin. Reprod. Med. 33, 410421.
Wood, T., Kelly, C., Roberts, M., Walsh, B., 2019. An interpretable machine learning
model of biological age. F1000Res. 8, 17.
World Health Organization, 2011. ICD-11 alpha. Content model reference guide. 11th
Revision World Health Organization, Geneva.
World Health Organization, 2010. Global recommendations on physical activity for
health. World Health Organization, Geneva.
Wu, C.-F., Liu, P.-Y., Wu, T.-J., Hung, Y., Yang, S.-P., Lin, G.-M., 2015a. Therapeutic
modication of arterial stiness: An update and comprehensive review. World J.
Cardiol. 7, 742.
Wu, H., Xia, Y., Jiang, J., Du, H., Guo, X., Liu, X., Li, C., Huang, G., Niu, K., 2015b. Eect
of beta-hydroxy-beta-methylbutyrate supplementation on muscle loss in older adults:
A systematic review and meta-analysis. Arch. Gerontol. Geriatr. 61, 168175.
Wu, P., Dugoua, J.J., Eyawo, O., Mills, E.J., 2009. Traditional Chinese Medicines in the
treatment of hepatocellular cancers: a systematic review and meta-analysis. J. Exp.
Clin. Cancer Res. 28, 112.
Wu, Y., Zhang, Q., Ren, Y., Ruan, Z., 2017. Eect of probiotic Lactobacillus on lipid
prole: A systematic review and meta-analysis of randomized, controlled trials. PLoS
One 12, e0178868.
Xia, Z., Cholewa, J., Zhao, Y., Shang, H.-Y., Yang, Y.-Q., Araújo Pessôa, K., Su, Q.-S.,
Lima-Soares, F., Zanchi, N.E., 2017. Targeting Inammation and Downstream Protein
Metabolism in Sarcopenia: A Brief Up-Dated Description of Concurrent Exercise and
Leucine-Based Multimodal Intervention. Front. Physiol. 8, 434.
Xie, W., Zhou, J., 2018. Aberrant regulation of autophagy in mammalian diseases. Biol.
Lett. 14. https://doi.org/10.1098/rsbl.2017.0540.
Xie, X., Atkins, E., Lv, J., Bennett, A., Neal, B., Ninomiya, T., Woodward, M., MacMahon,
S., Turnbull, F., Hillis, G.S., Chalmers, J., Mant, J., Salam, A., Rahimi, K., Perkovic,
V., Rodgers, A., 2016. Eects of intensive blood pressure lowering on cardiovascular
and renal outcomes: updated systematic review and meta-analysis. Lancet 387,
435443.
Xu, B., Xie, X., 2016. Neurotrophic factor control of satiety and body weight. Nat. Rev.
Neurosci. 17, 282292.
Xu, B., Zhou, X., Li, X., Liu, C., Yang, C., 2017. Diabetes mellitus carries a risk of eso-
phageal cancer: A meta-analysis. Medicine 96, e7944.
Xu, X., Qu, K., Pang, Q., Wang, Z., Zhou, Y., Liu, C., 2016. Association between telomere
length and survival in cancer patients: a meta-analysis and review of literature. Front.
Med. 10, 191203.
Yamagishi, S.-I., Nakamura, N., Suematsu, M., Kaseda, K., Matsui, T., 2015. Advanced
Glycation End Products: A Molecular Target for Vascular Complications in Diabetes.
Mol. Med. 21 (Suppl 1), S3240.
Yang, L., Ling, W., Du, Z., Chen, Y., Li, D., Deng, S., Liu, Z., Yang, L., 2017. Eects of
Anthocyanins on Cardiometabolic Health: A Systematic Review and Meta-Analysis of
Randomized Controlled Trials. Adv. Nutr. 8, 684693.
Yang, L., Lof, M., Veierød, M.B., Sandin, S., Adami, H.-O., Weiderpass, E., 2011.
Ultraviolet exposure and mortality among women in Sweden. Cancer Epidemiol.
Biomarkers Prev. 20, 683690.
Yaniv, Y., Juhaszova, M., Sollott, S.J., 2013. Age-related changes of myocardial ATP
supply and demand mechanisms. Trends Endocrinol. Metab. 24, 495.
Yashin, A.I., Arbeev, K.G., Akushevich, I., Arbeeva, L., Kravchenko, J., Ilyasova, D.,
Kulminski, A., Akushevich, L., Culminskaya, I., Wu, D., Ukraintseva, S.V., 2010.
Dynamic Determinants of Longevity and Exceptional Health. Curr. Gerontol. Geriatr.
Res. https://doi.org/10.1155/2010/381637. 2010.
Ye, W., Zhu, S., Liao, C., Xiao, J., Wu, Q., Lin, Z., Chen, J., 2017. Advanced oxidation
protein products induce apoptosis of human chondrocyte through reactive oxygen
species-mediated mitochondrial dysfunction and endoplasmic reticulum stress path-
ways. Fundam. Clin. Pharmacol. 31, 6474.
Yin, F., Sancheti, H., Patil, I., Cadenas, E., 2016. Energy metabolism and inammation in
brain aging and Alzheimers disease. Free Radic. Biol. Med. 100, 108122.
Ying, W., 2008. NAD+/NADH and NADP+/NADPH in cellular functions and cell death:
regulation and biological consequences. Antioxid. Redox Signal. 10, 179206.
Yip, W.C.Y., Sequeira, I.R., Plank, L.D., Poppitt, S.D., 2017. Prevalence of Pre-Diabetes
across Ethnicities: A Review of Impaired Fasting Glucose (IFG) and Impaired Glucose
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
26
Tolerance (IGT) for Classication of Dysglycaemia. Nutrients 9. https://doi.org/10.
3390/nu9111273.
Yoshino, J., Baur, J.A., Imai, S.-I., 2018. NAD+ Intermediates: The Biology and
Therapeutic Potential of NMN and NR. Cell Metab. 27, 513528.
Yu, H.T., Youn, J.-C., Kim, J.H., Seong, Y.-J., Park, S.-H., Kim, H.C., Lee, W.-W., Park, S.,
Shin, E.-C., 2017. Arterial Stiness Is Associated With Cytomegalovirus-Specic
Senescent CD8+ T Cells. J. Am. Heart Assoc. 6. https://doi.org/10.1161/JAHA.117.
006535.
Yu-Wai-Man, P., Lai-Cheong, J., Borthwick, G.M., He, L., Taylor, G.A., Greaves, L.C.,
Taylor, R.W., Griths, P.G., Turnbull, D.M., 2010. Somatic mitochondrial DNA de-
letions accumulate to high levels in aging human extraocular muscles. Invest.
Ophthalmol. Vis. Sci. 51, 33473353.
Zaccardi, F., ODonovan, G., Webb, D.R., Yates, T., Kurl, S., Khunti, K., Davies, M.J.,
Laukkanen, J.A., 2015. Cardiorespiratory tness and risk of type 2 diabetes mellitus:
A 23-year cohort study and a meta-analysis of prospective studies. Atherosclerosis
243, 131137.
Zaslavsky, O., Walker, R.L., Crane, P.K., Gray, S.L., Larson, E.B., 2016. Glucose Levels and
Risk of Frailty. J. Gerontol. A Biol. Sci. Med. Sci. 71, 12231229.
Zhai, J., Bo, Y., Lu, Y., Liu, C., Zhang, L., 2017. Eects of Coenzyme Q10 on Markers of
Inammation: A Systematic Review and Meta-Analysis. PLoS One 12. https://doi.
org/10.1371/journal.pone.0170172.
Zhang, G., Li, J., Purkayastha, S., Tang, Y., Zhang, H., Yin, Y., Li, B., Liu, G., Cai, D., 2013.
Hypothalamic programming of systemic ageing involving IKK-β, NF-κB and GnRH.
Nature 497, 211216.
Zhang, H., Menzies, K.J., Auwerx, J., 2018a. The role of mitochondria in stem cell fate
and aging. Development 145. https://doi.org/10.1242/dev.143420.
Zhang, D., Li, N., Xi, Y., Zhao, Y., Wang, T., 2017. Diabetes mellitus and risk of ovarian
cancer. A systematic review and meta-analysis of 15 cohort studies. Diabetes Res.
Clin. Pract. 130, 4352.
Zhang, Q., Liu, J., Liu, J., Huang, W., Tian, L., Quan, J., Wang, Y., Niu, R., 2014a. oxLDL
induces injury and defenestration of human liver sinusoidal endothelial cells via
LOX1. J. Mol. Endocrinol. 53, 281293.
Zhang, Q., Wu, Y., Fei, X., 2015. Eect of probiotics on body weight and body-mass index:
a systematic review and meta-analysis of randomized, controlled trials. Int. J. Food
Sci. Nutr. 67, 571580.
Zhang, W.-G., Zhu, S.-Y., Bai, X.-J., Zhao, D.-L., Jian, S.-M., Li, J., Li, Z.-X., Fu, B., Cai, G.-
Y., Sun, X.-F., Chen, X.-M., 2014b. Select aging biomarkers based on telomere length
and chronological age to build a biological age equation. Age 36, 9639.
Zhang, Y., Feng, B., 2017. Systematic review and meta-analysis for the association of bone
mineral density and osteoporosis/osteopenia with vascular calcication in women.
Int. J. Rheum. Dis. 20, 154160.
Zhang, Y., Ma, K.L., Ruan, X.Z., Liu, B.C., 2016. Dysregulation of the Low-Density
Lipoprotein Receptor Pathway Is Involved in Lipid Disorder-Mediated Organ Injury.
Int. J. Biol. Sci. 12, 569579.
Zhang, Y., Qi, L., Xu, L., Sun, X., Liu, W., Zhou, S., van de Vosse, F., Greenwald, S.E.,
2018b. Eects of exercise modalities on central hemodynamics, arterial stiness and
cardiac function in cardiovascular disease: Systematic review and meta-analysis of
randomized controlled trials. PLoS One 13, e0200829.
Zhang, Y., Zhang, D.-Z., 2018. Is coee consumption associated with a lower level of
serum C-reactive protein? A meta-analysis of observational studies. Int. J. Food Sci.
Nutr. 69, 985994.
Zhao, H., Song, A., Zhang, Y., Shu, L., Song, G., Ma, H., 2019a. Eect of Resveratrol on
Blood Lipid Levels in Patients with Type 2 Diabetes: A Systematic Review and Meta-
Analysis. Obesity 27, 94102.
Zhao, W., Ammous, F., Ratli, S., Liu, J., Yu, M., Mosley, T.H., Kardia, S.L.R., Smith, J.A.,
2019b. Education and Lifestyle Factors Are Associated with DNA Methylation Clocks
in Older African Americans. Int. J. Environ. Res. Public Health 16. https://doi.org/
10.3390/ijerph16173141.
Zhao, Y., Wu, K., Zheng, J., Zuo, R., Li, D., 2015. Association of coee drinking with all-
cause mortality: a systematic review and meta-analysis. Public Health Nutr. 18,
12821291.
Zhavoronkov, A., Bhullar, B., 2015. Classifying aging as a disease in the context of ICD-
11. Front. Genet. 6, 326.
Zheng, Y., Luo, X., Zhu, J., Zhang, X., Zhu, Y., Cheng, H., Xia, Z., Su, N., Zhang, N., Zhou,
J., 2012. Mitochondrial DNA 4977 bp deletion is a common phenomenon in hair and
increases with age. Bosn. J. Basic Med. Sci. 12, 187.
Zhou, D., Yin, D., Xiao, F., Hao, J., 2015. Expressions of Senescence-Associated β-
Galactosidase and Senescence Marker Protein-30 are Associated with Lens Epithelial
Cell Apoptosis. Med. Sci. Monit. 21, 37283735.
Zhou, X., Li, Y., Shi, X., Ma, C., 2016. An overview on therapeutics attenuating amyloid β
level in Alzheimers disease: targeting neurotransmission, inammation, oxidative
stress and enhanced cholesterol levels. Am. J. Transl. Res. 8, 246.
Zhu, X.-H., Lu, M., Lee, B.-Y., Ugurbil, K., Chen, W., 2015. In vivo NAD assay reveals the
intracellular NAD contents and redox state in healthy human brain and their age
dependences. Proc. Natl. Acad. Sci. U. S. A. 112, 28762881.
Zubakov, D., Liu, F., van Zelm, M.C., Vermeulen, J., Oostra, B.A., van Duijn, C.M.,
Driessen, G.J., van Dongen, J.J.M., Kayser, M., Langerak, A.W., 2010. Estimating
human age from T-cell DNA rearrangements. Curr. Biol. 20 R9701.
D. Khaltourina, et al. Mechanisms of Ageing and Development 189 (2020) 111230
27
... For this reason, some researchers proposed to the World Health Organization (WHO) that aging and old age be recognized and included as a disease in the International Classification of Diseases 11th Revision (ICD-11). The proposal was approved in 2019, to enter into force in 2022 [4][5][6]; however, said approval generated broad rejection in the geriatric and gerontological field, because it was included in the term "Old age" under general symptoms (code MG2A), the WHO responded to this claim by replacing the term "Old age" with "Ageing associated decline in intrinsic capacity" [7][8][9]. However, although the proposal is qualified with said change, the codes XT9T (Ageing-related) and MG2A (Ageing-associated decline in intrinsic capacity) are maintained in the recently published ICD-11 [10], for which reason the WHO currently considers aging as a disease. ...
... Calimport and Bentley (2019) highlighted the importance of the WHO classifying aging as a disease, supporting the "anti-aging" approach [5]. Likewise, Khaltourina et al. (2020) emphasized the achievement of the WHO recognizing aging and old age as diseases [6], however, the claim of several geriatricians and gerontologists has been pointed out, which led to its exclusion at the same time in the ICD-11 as a disease [7][8][9]. However, it is very disconcerting and unheard of that the WHO maintains aging as a disease in its classification [10], which can be classified as a hallmark of institutional ageism. ...
... Calimport and Bentley (2019) highlighted the importance of the WHO classifying aging as a disease, supporting the "anti-aging" approach [5]. Likewise, Khaltourina et al. (2020) emphasized the achievement of the WHO recognizing aging and old age as diseases [6], however, the claim of several geriatricians and gerontologists has been pointed out, which led to its exclusion at the same time in the ICD-11 as a disease [7][8][9]. However, it is very disconcerting and unheard of that the WHO maintains aging as a disease in its classification [10], which can be classified as a hallmark of institutional ageism. ...
Article
Full-text available
Ageism is a type of discrimination characterized by negative social representations of old age and aging, with prejudices and stereotypes that cause rejection and marginalization of older adults, generally considering them as fragile and unproductive. For this reason, it is recognized as one of the main enemies of healthy aging, especially when it arises from the scientific and professional fields. In this sense, the proposals promoted by some researchers regarding the World Health Organization (WHO) classifying aging as a disease goes against the healthy aging approach. In this sense, we consider that there is no theoretical or scientific support to classify aging as a disease, so we must advocate before the WHO so that aging is eliminated within its disease classification codes. In this framework, this review proposes the concept of "hallmarks of ageism” defined as the characteristics, representations and attitudes of rejection and discrimination towards aging, old age and older people, at the political and institutional, scientific or professional, technological and digital, social, family and personal levels, which are presented in an articulated and structured manner. For this reason, it is essential to comprehensively identify and analyze the “hallmarks of ageism”, in order to propose programs that include strategies and public policies that promote “anti-ageism” as a counterproposal to the "hallmarks of aging", whose biological changes related to aging are intended to be comparable to chronic non-communicable diseases.
... Aging is a natural aspect of biological progression during which body function gradually diminishes, making the body increasingly susceptible to various diseases and health concerns. It significantly contributes to the risk of age-related conditions, such as heart problems, brain-related disorders, cancer, declining bone health, and diminished muscle strength [3,4]. ...
Article
Full-text available
Pearl oysters have been extensively utilized in pearl production; however, most pearl oyster shells are discarded as industrial waste. In a previous study, we demonstrated that the intraperitoneal administration of pearl oyster shell-derived nacre extract (NE) prevented d-galactose-induced brain and skin aging. In this study, we examined the anti-aging effects of orally administered NE in senescence-accelerated mice (SAMP8). Feeding SAMP8 mice NE prevented the development of aging-related characteristics, such as coarse and dull hair, which are commonly observed in aged mice. Additionally, the NE mitigated muscle aging in SAMP8 mice, such as a decline in grip strength. Histological analysis of skeletal muscle revealed that the NE suppressed the expression of aging markers, cyclin-dependent kinase inhibitor 2A (p16) and cyclin-dependent kinase inhibitor 1 (p21), and increased the expression of sirtuin1 and peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1)- α, which are involved in muscle synthesis. These findings suggest that the oral administration of NE suppresses skeletal muscle aging. Moreover, NE administration suppressed skin aging, including a decline in water content. Interestingly, oral administration of NE significantly extended the lifespan of SAMP8 mice, suggesting that its effectiveness as an anti-aging agent of various tissues including skeletal muscle, skin, and adipose tissue.
... Aging, a complex network characterized by multiple changes occurring at different biological levels, is a pathological process that leads to the deterioration of cellular, tissue, and organismal functions [1][2] . Aging needs to meet ICD-11 criteria to be considered a disease [3] . Mammalian aging is often attributed to molecular crosslinking, free radical-induced damage, telomere shortening, and methylation of DNA [4][5][6][7] . ...
Article
Objective: To study the anti-aging effects of Radix Notoginseng and to explore its molecular network mechanism. Methods: Aging and Radix notoginseng gene targets were searched and downloaded from the Genecards website, then Venn intersection analysis was performed to find common genes for diseases and drugs to explore candidate targets for Radix notoginseng in the treatment of aging. Bioinformatics was then used to analyze the biological processes, cellular components, molecular functions, and KEGG signaling pathways of the shared target network. Protein molecular network construction was carried out to find the core molecular network genes of the drug Radix notoginseng for the treatment of aging. A final PubMed literature comparison was performed to assess the value of the potential role of core network genes. Results: The keywords “Aging” and “Radix notoginseng” were queried in Genecards and 25,000 aging-related targets were obtained, 17 for Radix notoginseng. GO and KEGG analysis of the intersecting genes obtained from the Venn intersection analysis then showed that the BP with the highest potential to be associated with disease and drugs is positive regulation of protein phosphorylation, CC is macromolecular complex and MF is identical protein binding. The KEGG with the higher correlation is lipid and atherosclerosis, AGE-RAGE signaling pathway in diabetic complications, and proteoglycans in cancer. A total of 10 hub genes were identified in the PPI network construction, including EGFR, MMP9, TNF, VEGFA, RHOA, CDKN1A, CASP3, CCND1, AKT1, and IL1B. Among these, it found that a large number of MMP9 and TNF genes were reported in the literature, with the remaining hub genes less frequently reported in the literature. Conclusion: This study uses bioinformatics and network pharmacology to explain the core network mechanisms of the drug Radix notoginseng in the treatment of aging using the latest databases. The results show that hub genes such as CDKN1A, EGFR, and AKT1 are involved in the core biological processes of aging. The results of the study provide an important reference for resolving the core molecular network mechanism of anti-aging properties and provide a validation basis for future experimental validation.
... Intervenções contra os processos patológicos relacionados ao envelhecimento incluem medidas de mudança do estilo de vida (dieta equilibrada, atividade física regular), suplementação com antioxidantes, medicamentos anti-inflamatórios (por exemplo, ácido acetilsalicílico, etc) e/ou específica para tratamento de doenças (por exemplo, metformina, pentoxifilina, etc) 7 . ...
Article
Aging can be related to a cognitive decline, as well as, poor functionality, frailty and may increase the risk of developing diseases such as: cancer, diabetes, cardiovascular diseases, musculoskeletal and/or neurodegenerative disorders - which contribute to a worse quality of life and increased risk of death. Nine factors can contribute to cell aging, which can be summarized in the mnemonic DESGASTAR. Cellular senescence contributes to immunosenescence, which generates a low degree of inflammation, mitochondrial dysfunction that contributes to the generation of reactive oxygen specimens (ROS). The reduction in glutathione levels can contribute to aging, in addition to promoting inflammation and/or production of free radicals, which can contribute to an increased risk of chronic degenerative diseases. Interventions that include lifestyle changes (balanced diet, regular exercise, no smoking and moderate or non-consumption of alcohol), in addition to amino acid, vitamin and or mineral supplementation, when medically indicated and needed, can delay the physiological process called age-associated cell decline.
... Factors like heavy lifting, chronic coughing, obesity, and menopausal hormonal shifts contribute significantly to its development. Recognizing these contributors is pivotal in formulating preventive strategies and targeted interventions (Khaltourina et al., 2020). Additionally, menopausal hormonal changes weaken pelvic support structures, exacerbating POP risk. ...
Article
Full-text available
Pelvic Organ Prolapse (POP) stands as a prevalent health concern among women worldwide, characterized by the descent or herniation of pelvic organs, including the bladder, uterus, and rectum, into the vaginal canal. Objective: The primary aim of the study is to find the occurrence of pelvic organ prolapse in women and its prevalence, contributing factors, and impact on quality of life. This cross-sectional study was conducted in Catchment areas of Rural Health Centres Kangra and Kot Najibullah, District Haripur, Pakistan, from 1st November 2022 to 31st November 2023. Data was collected from 3284 women to find the pelvic organ prolapse in women and its prevalence, contributing factors, and impact on quality of life. The study comprised females diagnosed with various stages and types of POP, confirmed through clinical examinations, imaging studies, and documented medical history. Data was collected from 3284 female patients. The mean age of the patients was 54.01 ± 7.61 years. 60% of females are in the Pre-menopausal stage and 40% in the Post-menopausal stage. In managing Pelvic Organ Prolapse (POP), treatment modalities varied in utilization and exhibited distinct outcomes. Non-surgical interventions were employed in 30% of cases, resulting in a notable 70% relief in symptoms and a high patient satisfaction rate of 65%. It is concluded that this is a substantial problem for the women affected, yet the majority of affected women did not seek medical care. Pelvic organ prolapse is highly prevalent in rural Pakistan, impacts women’s everyday lives, and remains mainly untreated.
Chapter
The title of this chapter expresses well not only its main idea, but also formulates one of the key ideas of the whole book. Grinin et al. give many arguments in favor of these ideas, and in the last paragraph of this chapter they summarize them. The authors believe that medicine of the future will be cardinally new kind of medicine because of anti-aging technologies, that is, technologies for slowing down and ennobling aging, as well as stopping and alleviating the problems associated with it. All this will become an integral part of the future of medicine; perhaps even reforming whole system of medicine. This will also mean that more and more attention will be focused on the treatment of age-associated diseases, on the cure or mitigating of the so-called geriatric syndromes (or age-related conditions). The perception of aging in society will also change. Thus, the changing focus of medicine on the problems of aging together with innovations in anti-aging technologies will imply a medical adaptation to aging. The latter will develop gradually up to the late twenty-first century and become an essential part of the common process that Grinin et al. have called adaptation to aging.
Chapter
Oxidative stress results from an imbalance between reactive oxygen species production and the capacity of the organism to counteract them. These species are reactive substances that may deteriorate lipids, proteins, and DNA, which causes cellular malfunction and acts as etiological factors for various age-related disorders, including aging. The gut microbiota contains trillions of microorganisms, including bacteria, protozoa, viruses, and fungi, as well as their combined genetic material, that are present in the gastrointestinal tract. These microbiomes can also influence oxidative stress through their metabolic activities. As we age, the gut microbiome undergoes changes in composition and diversity, which can alter its metabolic activities and contribute to oxidative stress through mechanisms like production of reactive oxygen species, and modulation of antioxidant defences. For example, certain gut bacteria can produce reactive oxygen species as a byproduct of their metabolism, while others can generate antioxidants that neutralise them. The balance between reactive oxygen species-producing and antioxidant-producing bacteria can shift as we age, leading to increased oxidative stress. In addition, the gut microbiome can influence the host’s antioxidant defence system by producing short-chain fatty acids and other metabolites that can modulate gene expression and signalling pathways. One of the short-chain fatty acids, butyrate, can result in an increase in the production of antioxidant enzymes such as catalase and superoxide dismutase and reduce oxidative stress in the gut and other organs. Additionally, available literature also indicates that oxidative stress can affect the gut microbiome by changing its structure and functionality through the immune response, which in turn influences the aging process. Oxidative byproducts can damage bacterial membranes, DNA, and proteins, leading to microbial cell death and dysbiosis. Dysbiosis, on the other hand, might increase oxidative stress by lowering the production of short-chain fatty acids and other antioxidant metabolites. Overall, the molecular interaction between oxidative stress and the gut microbiome in aging is intricate and complicated. Further research is required to understand the mechanisms underlying these interactions and identify strategies to promote a healthy gut microbiome and reduce oxidative stress in the aging process.
Chapter
Full-text available
The high prevalence of multiple comorbidities poses unique medication-related challenges for geriatric patients. Polypharmacy is a particular concern since taking several medications simultaneously increases the likelihood of adverse drug events and the risk of drug interactions while decreasing patient adherence. These factors are associated with suboptimal health outcomes and a heightened burden on the healthcare system (insurance claims) and the patient (out-of-pocket expenses). These challenges can significantly affect the quality of life of geriatric patients. This chapter critically examines the impact of medication use and polypharmacy on the quality of life of older patients. In addition, we discuss how artificial intelligence-based digital tools and precision medicine can address these issues by streamlining medical decision-making, improving the patient experience, and allowing remote monitoring. Finally, we interpret our findings from the lens of ethical considerations associated with the adoption and implementation of digital applications and gadgets.
Chapter
Full-text available
The causes of death reported on the death certificates of the oldest old are generally seen as unreliable, and as thus providing little useful information on the process leading to death. However, in advanced countries, a majority of the people who die each year are relatively old, and the level of detail provided on medical certificates about the causes of death among this older population is improving. At the same time, scholars are becoming increasingly interested in studying not just the initial cause of death, but multiple causes of death, thereby taking all of the information reported on the certificate into account. This study demonstrates that in a country like France, the cause-of-death pattern evolves regularly until around age 105. The share of people dying of circulatory diseases tends to be quite stable over the age range, while the share of individuals dying of cancer is declining, and the share of people dying of respiratory/infectious diseases is rising. Furthermore, among people who die at very old ages, a typology of multiple causes of death highlights the growing importance of ill-defined causes, while opening the door to an interesting discussion about the concept of cause of death in the supercentenarian population. Instead of representing an ill-defined cause, senility could be considered an actual cause of death. This suggests that daily care is more crucial to the survival of the oldest old than any conventional medical care or treatment. Supercentenarians tend to be so frail that any minor health event or brief lapse of attention on the part of their caregivers can be lethal.
Article
Full-text available
Aging is a predominant risk factor for several chronic diseases that limit healthspan¹. Mechanisms of aging are thus increasingly recognized as potential therapeutic targets. Blood from young mice reverses aspects of aging and disease across multiple tissues2–10, which supports a hypothesis that age-related molecular changes in blood could provide new insights into age-related disease biology. We measured 2,925 plasma proteins from 4,263 young adults to nonagenarians (18–95 years old) and developed a new bioinformatics approach that uncovered marked non-linear alterations in the human plasma proteome with age. Waves of changes in the proteome in the fourth, seventh and eighth decades of life reflected distinct biological pathways and revealed differential associations with the genome and proteome of age-related diseases and phenotypic traits. This new approach to the study of aging led to the identification of unexpected signatures and pathways that might offer potential targets for age-related diseases.
Article
Full-text available
Much of the key epidemiological evidence that long-term exposure to fine particulate matter air pollution (PM2.5) contributes to increased risk of mortality comes from survival studies of cohorts of individuals. Although the first two of these studies, published in the mid-1990s, were highly controversial, much has changed in the last 25 + years. The objectives of this paper are to succinctly compile and summarize the findings of these cohort studies using meta-analytic tools and to address several of the key controversies. Independent reanalysis and substantial extended analysis of the original cohort studies have been conducted and many additional studies using a wide variety of cohorts, including cohorts constructed from public data and leveraging natural experiments have been published. Meta-analytic estimates of the mean of the distribution of effects from cohort studies that are currently available, provide substantial evidence of adverse air pollution associations with all-cause, cardiopulmonary, and lung cancer mortality.
Article
Full-text available
Globally, citizens exist for sustained periods in states of aging-related disease and multimorbidity. Given the urgent and unmet clinical, health care, workforce, and economic needs of aging populations, we need interventions and programs that regenerate tissues and organs and prevent and reverse aging-related damage, disease, and frailty (1). In response to these challenges, the World Health Organization (WHO) has called for a comprehensive public-health response within an international legal framework based on human rights law (1). Yet for a clinical trial to be conducted, a disease to be diagnosed, intervention prescribed, and treatment administered; a corresponding disease classification code is needed, adopted nationally from the WHO International Classification of Diseases (ICD). Such classifications and staging are fundamental for health care governance among governments and intergovernmental bodies. We describe a systematic and comprehensive approach to the classification and staging of organismal senescence and aging-related diseases at the organ and tissue levels in order to guide policy and practice and enable appropriate interventions and clinical guidance, systems, resources, and infrastructure.
Article
Full-text available
Epigenetic "clocks" can now surpass chronological age in accuracy for estimating biological age. Here, we use four such age estimators to show that epigenetic aging can be reversed in humans. Using a protocol intended to regenerate the thymus, we observed protective immunological changes, improved risk indices for many age-related diseases, and a mean epigenetic age approximately 1.5 years less than baseline after 1 year of treatment (-2.5-year change compared to no treatment at the end of the study). The rate of epigenetic aging reversal relative to chronological age accelerated from -1.6 year/year from 0-9 month to -6.5 year/year from 9-12 month. The GrimAge predictor of human morbidity and mortality showed a 2-year decrease in epigenetic vs. chronological age that persisted six months after discontinuing treatment. This is to our knowledge the first report of an increase, based on an epigenetic age estimator, in predicted human lifespan by means of a currently accessible aging intervention.
Article
Full-text available
DNA methylation (DNAm) clocks are important biomarkers of cellular aging and are associated with a variety of age-related chronic diseases and all-cause mortality. Examining the relationship between education and lifestyle risk factors for age-related diseases and multiple DNAm clocks can increase the understanding of how risk factors contribute to aging at the cellular level. This study explored the association between education or lifestyle risk factors for age-related diseases and the acceleration of four DNAm clocks, including intrinsic (IEAA) and extrinsic epigenetic age acceleration (EEAA), PhenoAge acceleration (PhenoAA), and GrimAge acceleration (GrimAA) in the African American participants of the Genetic Epidemiology Network of Arteriopathy. We performed both cross-sectional and longitudinal analyses. In cross-sectional analyses, gender, education, BMI, smoking, and alcohol consumption were all independently associated with GrimAA, whereas only some of them were associated with other clocks. The effect of smoking and education on GrimAA varied by gender. Longitudinal analyses suggest that age and BMI continued to increase GrimAA, and that age and current smoking continued to increase PhenoAA after controlling DNAm clocks at baseline. In conclusion, education and common lifestyle risk factors were associated with multiple DNAm clocks. However, the association with each risk factor varied by clock, which suggests that different clocks may capture adverse effects from different environmental stimuli.
Article
Background: Although epidemiology studies have reported the relationship, including a dose-response relationship, between dietary magnesium intake and risk of cardiovascular disease (CVD), the risk for CVD mortality is inconclusive and the evidence for a dose-response relationship has not been summarized. Objective: We conducted a systematic review and meta-analysis of prospective studies to summarize the evidence regarding the association of dietary magnesium intake with risk of CVD mortality and describe their dose-response relationship. Design: We identified relevant studies by searching major scientific literature databases and grey literature resources from their inception to August 2015, and reviewed references lists of retrieved articles. We included population-based studies that reported mortality risks, i.e. relative risks (RRs), odds ratios (ORs) or hazard ratios (HRs) of CVD mortality or cause-specific CVD death. Linear dose-response relationships were assessed using random-effects meta-regression. Potential nonlinear associations were evaluated using restricted cubic splines. Results: Out of 3002 articles, 9 articles from 8 independent studies met the eligibility criteria. These studies comprised 449,748 individuals and 10,313 CVD deaths. Compared with the lowest dietary magnesium consumption group in the population, the risk of CVD mortality was reduced by 16% in women and 8% in men. No significant linear dose-response relationship was found between increment in dietary magnesium intake and CVD mortality across all the studies. After adjusting for age and BMI, the risk of CVD mortality was reduced by 24-25% per 100mg/d increment in dietary magnesium intake in women of all the participants and in all the US participants. Conclusion: Although the combined data confirm the role of dietary magnesium intake in reducing CVD mortality, the dose-response relationship was only found among women and in US population.
Article
Importance Previous studies have reported that drug treatments, particularly treatment with bisphosphonates, is associated with reduced overall mortality rates in addition to decreased fracture risk. If so, drug treatments should be recommended for this reason alone, regardless of a patient’s risk of fracture. Objective To assess whether randomized clinical trials demonstrate that treatment with bisphosphonates, particularly zoledronate, is associated with reduced mortality rates. Data Sources Science Direct, MEDLINE, Embase, and the Cochrane Library were searched for randomized placebo-controlled clinical trials of drug treatments for osteoporosis published after 2009 and published or in press before April 19, 2019. Conference abstracts from annual osteoporosis society meetings were also included in the search. Study Selection Included studies were clinical trials that (1) were randomized and placebo-controlled; (2) studied drug treatments with proven antifracture efficacy; (3) used agents at the approved dose for treatment of osteoporosis; and (4) had a duration of 1 year or more. Abstracts from the literature searches were reviewed for inclusion and exclusion criteria, and mortality rate data were abstracted from the article by 1 researcher and validated by a second. A total of 2045 records were screened; 38 (1.8%) were included in the meta-analyses. Data Extraction and Synthesis The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) checklist was followed for abstracting data and assessing data quality and validity. Data were pooled using random-effects models, and between-study variability was assessed using the I² index. The risk of bias for each study was assessed, and funnel plots and Egger and Begg statistics were used to evaluate publication bias. Main Outcomes and Measures Associations of all drug treatments, particularly bisphosphonate and zoledronate treatments, with overall mortality. Results Of 38 clinical trials that included 101 642 unique participants, 38 were included in the meta-analyses of all drug treatments (45 594 participants randomized to placebo; 56 048 to treatment); 21 clinical trials, of bisphosphonate treatments (20 244 participants randomized to placebo; 22 623 to treatment); and 6 clinical trials, of zoledronate treatments (6944 participants randomized to placebo; 6926 to treatment). No significant association was found between all drug treatments for osteoporosis and overall mortality rate (risk ratio [RR], 0.98; 95% CI, 0.91-1.05; I² = 0%). Clinical trials of bisphosphonate treatment (RR, 0.95; 95% CI, 0.86-1.04) showed no significant association with overall mortality. Also, clinical trials of zoledronate treatment (RR, 0.88; 95% CI, 0.68-1.13) showed no association with overall mortality rate; however, evidence existed for heterogeneity of the results (I² = 48.2%). Conclusions and Relevance Results of this meta-analysis suggest that bisphosphonate treatment may not be associated with reduced overall mortality rates in addition to decreased fracture risk and should only be recommended to reduce fracture risk. Additional trials are needed to clarify whether treatment with zoledronate reduces mortality rates.