Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Sep 19.
Published in final edited form as: Vet Pathol. 2016 Feb 11;53(2):277–290. doi: 10.1177/0300985815622974

Contributions of Nonhuman Primates to Research on Aging

E S Didier 1, A G MacLean 2, M Mohan 2, P J Didier 2, A A Lackner 2, M J Kuroda 3
PMCID: PMC5027759  NIHMSID: NIHMS815365  PMID: 26869153

Abstract

Aging is the biological process of declining physiologic function associated with increasing mortality rate during advancing age. Humans and higher nonhuman primates exhibit unusually longer average life spans as compared with mammals of similar body mass. Furthermore, the population of humans worldwide is growing older as a result of improvements in public health, social services, and health care systems. Comparative studies among a wide range of organisms that include nonhuman primates contribute greatly to our understanding about the basic mechanisms of aging. Based on their genetic and physiologic relatedness to humans, nonhuman primates are especially important for better understanding processes of aging unique to primates, as well as for testing intervention strategies to improve healthy aging and to treat diseases and disabilities in older people. Rhesus and cynomolgus macaques are the predominant monkeys used in studies on aging, but research with lower nonhuman primate species is increasing. One of the priority topics of research about aging in nonhuman primates involves neurologic changes associated with cognitive decline and neurodegenerative diseases. Additional areas of research include osteoporosis, reproductive decline, caloric restriction, and their mimetics, as well as immune senescence and chronic inflammation that affect vaccine efficacy and resistance to infections and cancer. The purpose of this review is to highlight the findings from nonhuman primate research that contribute to our understanding about aging and health span in humans.

Keywords: nonhuman primates, aging, animal models, inflammation, gerontology, frailty, senescence, caloric restriction, cognitive decline, immune senescence, Parkinson disease, Alzheimer disease, reproductive senescence, osteoporosis


Nonhuman primates share similar physiology and a close phylogenetic relationship to humans. The use of nonhuman primates in comparative experimental studies thus contributes to our knowledge about aging processes and translation of applications for improving health span in humans and other animals. Key evolutionary and adaptive features in nonhuman primates include pentadactylism for grasping and manipulating objects, binocular vision enabling 3-dimensional depth perception that is related to development of the cerebral cortex, and progression toward bipedalism and upright posture.124,224 Correlates of average natural life span of an organism are highly complex, but body size in conjunction with metabolism, reproduction, immunity, and environmental stress, among other factors, is associated with average longevity such that larger animal species tend to live longer.14,63 Interestingly, human and nonhuman primates exhibit unusually longer average life spans that are nearly 4-fold higher than those of most other mammals relative to their body sizes.14 In addition, nonhuman primates exhibit similar key life span metrics as humans, such as higher infant mortality rate, followed by lower mortality during the juvenile stage and then an extended period of increasing age-related morbidity and mortality.32 Since nonhuman primates can be studied under well-controlled experimental conditions, intervention strategies for evaluating vaccines or drugs are often tested in nonhuman primates for safety and efficacy before application in humans. This review highlights the contributions of nonhuman primate research to our understanding about aging and health span in humans.

By far, the predominant nonhuman primate species utilized in biomedical research facilities as well as for studies on aging are rhesus macaques (Macaca mulatta) and cynomolgus macaques (Macaca fasicularis).124,128 Specifically, among the facilities with nonhuman primates in North America that were recently surveyed, 80% housed rhesus macaques of Indian or Chinese origin, followed closely by cynomolgus macaques housed in 73% of the facilities.128 Aspects of aging research studies that utilize macaques include neurobiology, anatomy, physiology, cognition, and behavior,18,163,184,212,216 as well as reproductive senescence,11 caloric restriction (CR),46,143 and immune senescence.10,148,213 The use of macaques in research appears to represent the best compromise between phylogenetic and physiologic relatedness to humans, cost efficiency, life span, resources, expertise in animal husbandry practices, and adaptability for translation of results to humans. To improve efficiency, accessibility, and applicability, however, increasing emphasis is being placed on purpose-bred animals and further advancing animal husbandry practices so that lower primates also may be included for relevant model development of research on aging.*

Prosimians, or “premonkeys,” are the most phylogenetically distant nonhuman primates from humans. Among the prosimians, grey mouse lemurs (Microcebus murinus) have been the most extensively studied for relating processes of aging in relation to humans.16,126 For example, the mouse lemur was the first nonhuman primate species to demonstrate a relationship between cerebral atrophy and cognitive decline with aging that simulated what was seen in aging humans.167 Neuroscience studies about memory, behavior, and psychomotor function have utilized both captive and wild mouse lemurs. Results from studies using mouse lemurs also demonstrated correlations between aging and renal function,5,142 sleep deprivation,174 circadian rhythms,84 reproduction,33,56 thermoregulation,202205 metabolism and dietary restriction,57,58,125,140,141,168 oxidative stress,53 and replicative senescence.197 The use of prosimians in research is more cost-efficient, but limitations include their smaller size that restricts specimen sampling; differences in metabolic, biochemical, and endocrine responses compared with humans; and a need for continued development in animal husbandry techniques to reduce stress-related behaviors of captive prosimians.16

Among the New World monkeys (NWMs), common marmosets (Callithrix jacchus) often are used to examine aging related to neurogenesis, vision, neuroanatomy, cognition, and pathogenesis of neurodegenerative diseases such as Parkinson and Alzheimer. Research on reproductive senescence132,179 and metabolic changes associated with aging (eg, insulin-resistance, diabetes, and obesity) also utilize common marmosets.159,178,200 Cotton-top tamarins (Saquinus oedipus) and squirrel monkeys (Saimiri sciureus) are used to examine reproductive senescence and in comparative studies of aging.§

Chimpanzees (Pan troglodytes) have been the most common great apes used in biomedical research, as they closely resemble humans structurally and genetically, with >98% DNA sequence homology.49 Chimpanzee use in research, however, has declined due to ethical concerns, high costs, and complexities in maintenance.7577

Psychosocial Influences

Quality of life during aging is dependent on complex interrelationships between psychosocial and physical health parameters. Studies of humans indicate that psychosocial dimensions comprise networks of social relationships, feelings of connectedness versus loneliness, and overall satisfaction with life that together compose predictors of longevity and quality of life.2 Measurements of such parameters often are subjective and utilize self-evaluations among humans of varying ages and socioeconomic strata. Analogous research about psychosocial influences and aging in nonhuman primates examines social rankings and stress in captive and free-ranging colonies. Rhesus macaques in free-ranging colonies exhibit a median life span of approximately 15 years of age due to predation, a lower ability of older animals to find food, reproductive stress with aging, and increased accumulation of “wear and tear” effects, also referred to as allostatic stress.101 Captive rhesus macaques, however, have an average median life span of 25 years. Comparison between the 2 housing conditions thus enables evaluation of environmental influences on restorative homeostatic mechanisms during aging.43,45,101

Social rank among aging females constitutes an important measure for relating psychosocial stress to aging that may be comparable to socioeconomic pressures and effects on quality of life in aging humans.2,101 Female social dominance ranking is inherited, and female rhesus macaques born to low-ranking females tend to remain at lower social status. Characteristics of the higher psychosocial stress associated with lower social status and aging have linked the hypothalamic-pituitary-adrenal axis, immune senescence, and reproductive stress with decreased longevity, allostatic (“wear and tear”) stress, and reduced quality of life.85,101 Measureable indicators of psychosocial stress and aging featured higher levels of fecal cortisol, increased proinflammatory serum cytokine levels, declining ex vivo natural killer cytotoxic activity (as a function of immune senescence), and declining body mass index and body weight. In addition, older lower-ranking female rhesus macaques exhibited reproductive stress and produced smaller infants at wider time intervals to reduce energy output than did higher-ranking older females.101 Reduced psychosocial stress during aging, however, was reported in rhesus macaques and Japanese macaques living in colonies where strong intergenerational relationships developed among females to presumably provide mutual support in conflicts with nonfamilial members.2,45,147

Caloric Restriction

Over 75 years ago, McCay et al reported that calorie intake reduction by approximately 30% of an ad libitum diet that avoided malnutrition increased longevity in rats.145 CR has since been reported to also extend average life span in many organisms, including yeast, worms, fruit flies, and mice.46,117 Beneficial effects in rhesus macaques suggest that CR also may contribute to improving human health span.46,117 Many of the basic mechanisms of longevity associated with CR that affect cell growth, metabolism, and reproduction are conserved across species, but physiologic outcomes and biomarkers of improving health span appear to be more complex in higher mammals and primates.148,229 In addition, CR may not be feasible in humans, because compliance and milestones would be difficult to monitor and achieve. For example, long-term CR in humans is associated with reduced libido and lower body temperature.193 Therefore, nonhuman primates are being used, first, to define the beneficial effects of CR that promote healthier aging and, second, to define and test pharmacologic CR mimetics for safety and efficacy that may then be applied to humans for improving health span and life span.46,103,105,117,131,141

Old World monkeys (OWMs), such as rhesus macaques, cynomolgus macaques, and baboons, play prominent roles in CR studies because they exhibit higher risks for obesity, insulin resistance, type 2 diabetes, immunologic decline, and metabolic syndrome related to cardiovascular disease (CVD) and impaired cognition, which also occur in aging humans.27,46,52,117,206,226 CR initiated in nonhuman primates after reaching maturity improved health span and was associated with increased insulin sensitivity, decreased hyperglycemia and incidence of type 2 diabetes, preservation of skeletal muscle function, reduced visceral white adipose fat, attenuated development of CVD, and reduced total energy expenditure. If initiated prior to adulthood, however, CR was deleterious to immune responses in rhesus macaques150 and, when administered to pregnant baboons, induced increasing insulin resistance and higher risk for developing type 2 diabetes in the offspring.40 The magnitude of increased life span as a consequence of CR, however, was reported to decline with increasing complexity of the organism.148,229 This contributes to a debate about whether rhesus macaques undergoing CR exhibit increased life span because of the varied or polymorphic genetic backgrounds in the test populations, different levels of physical activity, and/or medical interventions applied to monkeys that may not have been used in studies with lower animals.46,104,117,143,148

Antiaging compounds are being tested for efficacy in non-human primates, especially in rhesus macaques fed a high-fat/high-sucrose diet that induces many of the morbidities associated with aging, such as obesity, type 2 diabetes, and CVD. Sirtuins are NAD+-dependent deacetylases that participate in DNA repair, mitochondrial biogenesis, and regulation of inflammation.131,154 Treatment of animals subjected to high-fat/high-sucrose diet with the sirtuin activator resveratrol prevented central arterial stiffening, increased insulin sensitivity, reduced blood glucose levels, preserved b cells in islets of Langerhans, reduced adipose cell size, and reduced macrophage infiltration with declining NFkB expression (ie, lowered inflammation).78,109,144 Mouse lemurs, photoperiod-dependent prosimian primates, also have been examined for effects of CR or resveratrol treatment as a CR mimetic. These animals acclimated well to their treatments after 1 year, and while oxidative stress–producing DNA and RNA damage increased initially, animals in the CR treatment group and in the resveratrol-supplemented diet subsequently exhibited reduced DNA and RNA damage during aging.58,141 The use of nonhuman primates is thus expected to continue for preclinical safety and efficacy testing of antiaging compounds.

Organ Systems

Immune System and Inflammation

Immune senescence is a decline in regulated immune responses and increasing susceptibility to infections as well as noncommunicable diseases. Chronic inflammation is one of the hallmarks of immune senescence associated with most diseases of aging and is commonly referred to as “inflammaging.”79,80 Advantages to using nonhuman primates to study immune senescence include similarities to humans in immune responses, cross-reactivity in immunologic reagents (eg, antibodies), and comparable range of infectious and noncommunicable diseases of aging, such as adenocarcinoma of the colon and chronic inflammation observed in rhesus macaques (Suppl. Figs. 1, 2). Also, monkeys can be naturally or experimentally infected with many of the same pathogens that infect humans to test efficacy of vaccines and treatments under controlled experimental conditions. Furthermore, nonhuman primates are amenable for use in longitudinal studies that require repeated sampling, and such studies can be better controlled for time of infection, compliance, and risk behaviors (eg, illicit drug use, cigarette smoking) that may confound interpretation of results in human studies.

Several reviews describing immune senescence in rhesus macaques have been published recently.91,149,152 Adaptive immune responses appear to be more affected with age than innate immune responses.91 A recent report comparing young, adult, and aged rhesus macaques, however, identified subtle increases in CD14+CD16+ monocytes (toward a nonclassical monocyte/macrophage phenotype) and a shift toward the CD11c+ myeloid dendritic cell population.10 Antigen-presenting cell populations and Toll-like receptor expression are broadly similar between adult and aged populations. A decline in ex vivo cytotoxic function in natural killer cells strongly correlates with aging and is predictive for impending mortality in aged rhesus macaques.44 Changing levels of circulating proinflammatory cytokines, such as increases in TNF-α, IL-6, and IFN-γ, also correlate with age in humans and nonhuman primates, especially rhesus macaques. It is still difficult, however, to specifically define the effects of shifting levels of cytokines, chemokines, and other circulating factors on biological versus chronological aging outcomes among individuals (ie, as measures of healthy vs less healthy aging).10,66,152

The most discernible changes during aging and in the adaptive immune system are observed in the T-lymphocyte compartment. Absolute numbers of circulating T cells do not change, but aging is associated with declining naïve T cells, increasing numbers of terminally differentiated effector memory T cells, and declining CD4+-to-CD8+ T cell ratios.66,91,106 The shifts in these cell populations are associated with decreasing progenitor stem cells in bone marrow and thymic atrophy, as also observed in nonhuman primates (Suppl. Figs. 3, 4). In addition, continued exposure to pathogens and persistent infections promote a shift toward increased effector memory T cells and a shrinking naïve T-cell antigen recognition repertoire.41

The repertoire and number of circulating B cells decline with aging in humans and rhesus macaques.66,91 Homeostatic responses or shifts in subpopulations of B cells, however, have not been fully examined in aging nonhuman primates. Mucosal antibody responses become compromised with increasing age. IgA responses to cholera toxin/cholera toxoid were lower in intestinal lavage specimens of older rhesus macaques, while IgM levels were higher.201 This was explained by the observation that emigration of IgA immunoblasts from Peyer patches to the small intestinal lamina propria declined in aging rhesus macaques due to reduced expression of homing molecules.183

Vaccine studies in older rhesus macaques further demonstrate declining immune competence with aging. Antibody responses in influenza-vaccinated rhesus macaques >19 years of age were significantly lower than in young adults aged 4 and 7 years, but boost immunizations in the older animals produced responses comparable to those of younger monkeys.42 Preexisting antibodies from earlier seasonal exposures are believed to explain why older humans were more resistant to the 2009 H1N1 influenza virus than younger individuals. Since humans could not be used to test this, naïve older adult rhesus macaques were infected with the CA04 H1N1 influenza virus. These animals developed higher viral loads than infected younger adults and also produced delayed T-cell responses, thereby supporting the hypothesis that preexisting antibodies in the older humans may be protective for some seasonal influenza viruses.112

Intervention strategies to improve immune responses during aging or to delay immune senescence are being tested in rhesus macaques as well. Older rhesus macaques vaccinated for influenza and treated with IL-7, a cytokine that promotes T-cell homeostasis, exhibited increased production of naïve T cells and higher antibody responses than older monkeys not given IL-7.9 CR similarly improves immune functions and delayed T-cell senescence in rhesus macaques when initiated during young adulthood (ie, approximately 5–7 years of age).143,151 Conversely, HIV-infected individuals undergoing antiretroviral therapy develop chronic diseases associated with aging earlier than non-HIV-infected persons and are considered to undergo accelerated aging.1,50,100 The rhesus macaque model of simian immunodeficiency virus (SIV) infection and antiretroviral therapy is emerging as a model to study immune response mechanisms of accelerated aging, as there are no reported genetic mutations in nonhuman primates analogous to Hutchinson-Gilford Progeria and Werner progeria syndromes that occur in humans.160

In summary, immune senescence and diseases of aging in humans and nonhuman primates are characterized by chronic inflammation and greater susceptibility to infectious diseases and malignancies. Responses to vaccines in the elderly are also reduced and less effective. These changes are generally associated with increasing levels of circulating proinflammatory cytokines and chemokines, a reduced naïve T-cell antigen recognition repertoire, and a growing population of effector memory T cells against persistent infections. It is anticipated that with aging of the human population worldwide and the growing pressures on public and medical health systems, the use of non-human primates will continue to expand for developing prevention and treatment strategies.

Central Nervous System

The effect of aging on the central nervous system is an area of intense interest in humans and thus contributes to the growth of studies examining similarities with nonhuman primates. As in humans, there is evidence of increased oxidative stress in aged rhesus macaques,210 which is linked to higher circulating levels of homocysteine and cognitive decline.223 CR, conversely, appears to contribute to successful cognitive and neurologic aging in macaques through maintaining white matter volume,21 lower levels of neuroinflammation,221 cortisol,222 and iron,116 as well as decreased mortality.28

Published studies describing lesions and changes in the central nervous system during aging most commonly use species of nonhuman primates phylogenetically closer to humans (Table 1). As nonhuman primates age, some neuronal populations are more affected than others, such as the shifts in density of calcium binding protein expression on neurons.88,89 For example, the percentage of calbindin-positive inhibitory GABAergic (gamma-aminobutyric acid) neurons were reported to increase over age in rhesus macaques, while there were fewer parvalbumin-positive glutamic acid decarboxylase 67 neurons. The investigators speculated that this shift in binding receptor density on neurons is important for buffering calcium and regulating excitotoxic effects. Reduced cognitive capacity in aged macaques was also associated with breakdown of myelin sheaths164 and axon loss.181 Furthermore, a reduced organization in neuronal arrangement was observed in aged animals97,221 that was related to loss of white matter volume in key brain areas, including frontal lobe and cerebellum,139 possibly contributing to an anterior-to-posterior gradient decline in white matter integrity195 and loss of synapses and synaptophysin expression.92 Overall brain volume declines in all primate species during aging but occurs proportionally later in chimpanzees than in humans.39 Aged humans, however, have a higher prevalence of neurodegenerative diseases causing dementia, including Alzheimer and Parkinson diseases. One explanation is that the longer life span in humans compared with nonhuman primates provides time for greater deterioration of white matter prior to death.39

Table 1.

Frequency of Studies of Age-Related Brain Disease Characteristics in Nonhuman Primates and Humans.a

Relative Frequency of Published Studies
Human Great Apes Macaque Marmoset Mouse Lemur
Brain volume (decline) / aging +++ +++ + + +/−
Alzheimer disease +++++ +++++ ++ + +
Neurodegeneration ++++ ++++ + +/− +/−
Neuronal loss +++ +++ + +/− +/−
Glial activation ++ ++ +/− +/− +/−
Amyloid in brain +++++ +++++ ++ +/− +
Tau (activation) ++++ ++++ + +/− +/−
α-Synuclein activation +++ + +/− +/− +/−
Parkinson disease +++++ ++ ++ ++ +/−
Glutamate (decrease) ++ + + + +/−
Glial activation ++ +/− +/− +/− +/−
Dyskinesia ++++ + ++ + +/−
Dopa therapy ++++ + ++ + +/−
a

Search terms indicated on the table were entered into PubMed (http://www.ncbi.nlm.nih.gov/pubmed), and the number of citations between 1964 through 2014 are indicated as follows: +/−, <10 citations; +, 10–99 citations; ++, 100–999 citations; +++, 1000–4999 citations; ++++, 5000 – 9999 citations; +++++, ≥ 10000 citations.

Alzheimer Disease

While all primates exhibit a degree of progressive age-related neurodegeneration, the drastic neuron loss and clinically observed cognitive decline of Alzheimer disease are considered unique to humans.77 Indeed, the great apes demonstrate fewer changes associated with brain aging than humans.77 Several nonhuman primate models, however, are being used to explore aspects of neurodegeneration that occur in Alzheimer disease.76,77,82,99,216

As noted above, diet is considered a major risk factor for metabolic syndrome and diabetes, which are associated with Alzheimer disease.121 Diet and obesity are linked to neuroinflammation (astrogliosis) in rhesus macaques but not to the presence of amyloid in the brain.196 However, in cynomolgus macaques, reactive astrocytes are reported to engulf β-amyloid,119 an important step in the initiation of amyloid plaque formation. This occurs in conjunction with lower Aβ42 in the cerebral spinal fluid and accumulation in the brain.227 Amyloid b deposits also are observed in brain of aged marmosets.82 Curiously, there are neither degenerative nor histologic changes, even in areas with plaques that were apparent by magnetic resonance imaging.211

Phosphorylation of tau protein is evident on glutamatergic synapses in aged rhesus macaques,34 and tau accumulation in cerebral spinal fluid is linked to poor memory in older cynomolgus macaques.60 Environmental influences or exposures during development may contribute to the increased phosphorylation and accumulation of tau protein that are associated with neurodegenerative disease later in life. For example, 23 years after being exposed to lead as infants, a group of rhesus macaques had greater levels of tau and β-amyloid in their brains than aged macaques that were not exposed to lead as infants.24 Neuropathology similar to that observed in human Alzheimer disease, such as tau hyperphosphorylation and extensive neurofibrillary degeneration, is seen in the mouse lemur as well.184 However, β-amyloid is rarely observed in nonhuman primate brains during aging,77,209 thus limiting the use of nonhuman primates in Alzheimer disease research. In this respect, the nonhuman primate brain may be a better model for normal brain aging as opposed to age-associated brain disease.

Parkinson Disease

Parkinson and Parkinson-like diseases are related to a loss of dopamine within synaptic vesicles169 or loss of these synapses.134 In rhesus and cynomolgus macaques, administration of 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP) is used to induce signs of Parkinson-like disease. MPTP administration activates microglia in rhesus and cynomolgus macaques19,114 and decreases glutamate levels in rhesus macaques.70 Rhesus and cynomolgus macaques also respond differently during the acute phase of MPTP administration. Cynomolgus macaques are generally more severely affected than rhesus macaques by measures of dyskinesia, parkinsonian motor disability scores, and functional recovery following withdrawal of MPTP.171 In addition, both species require multiple injections of MPTP to induce stable Parkinson-like disease, but the doses required vary among individual monkeys of both species.

Similar to humans, treatment with L-DOPA significantly improves the clinical neurologic function of cynomolgus macaques.87,156 In rhesus macaques, however, L-DOPA treatment produced limited recovery since hypokinesia and tremor were responsive to L-DOPA, but gait disorders did not improve.87 The comparatively better outcome in cynomolgus macaques may result from using antagonists to glutamatergic receptors in addition to L-DOPA to prevent dysregulation of associated signaling proteins, such as pERK, D2 receptors, and protein kinase B.156

The macaque models of MPTP-induced Parkinson-like disease also explored gene therapy approaches to correct for the loss of dopaminergic neurons.230 These studies used adenoviral vectors to transduce rhesus adipose mesenchymal stem cells to stably express neurturin and tyrosine hydroxylase. Once these stem cells differentiated into dopaminergic neurons and implanted into striatum and substantia nigra, there were significant improvements in the behavioral signs (eg, balance disorder scores) of MPTP toxicity in rhesus macaques. In addition to the MPTP toxicity model, a second model of Parkinson disease in marmosets used a recombinant adeno-associated virus vector to overexpress α-synuclein in dopaminergic neurons within the substantia nigra pars compacta. This approach was successful in inducing the loss of dopaminergic neurons and expression of Parkinson-like signs.74

Respiratory System

The lung undergoes changes in structure and function during aging that contribute to increased chronic lower respiratory tract diseases (eg, emphysema, bronchitis, chronic obstructive pulmonary disease [COPD]), which compose the third-leading cause of death in humans.137,186 Alterations in the ribs and spine, as well as declines in muscle strength and efficiency, contribute to decreased thoracic cavity volume as well as reduced maximum lung inhalation and expiration volumes. Furthermore, the oxidative stress from inflammatory responses to infectious agents and pollutants produces damage to lung tissue architecture and adversely affects efficiency of gas exchange. Changes observed in lung tissue of aged rhesus macaques, for example, demonstrated pigment accumulation in macrophages as well as atheromatous arterial thickening (Suppl. Figs. 5, 6).

COPD dramatically increases in prevalence in elderly humans and is associated with greater susceptibility to respiratory infections.186 Nonhuman primate models are used to better understand risk factors and mechanisms of COPD. For example, cigarette smoking is considered a principal risk factor in genetically susceptible individuals and exposure of cynomolgus macaques to cigarette smoke produced pulmonary disease similar to that observed in humans (eg, airway inflammation, peribronchial fibrosis, mucus metaplasia, bronchial lymphoid aggregates).170 HIV-infected individuals with Pneumocystis jiroveci (syn carinii) pneumonia develop COPD, and this is being modeled in rhesus macaques with SIV and Pneumocystis coinfection.120,190

Cardiovascular System

CVDs comprising coronary heart disease, heart failure, peripheral vascular disease, and stroke are the most common causes of death in older persons living in the developed world. Normal changes associated with aging include increased vascular rigidity or stiffening and reduced compliance or elasticity.129,158 These changes are exacerbated by comorbidities and risk factors, such as diabetes, hypertension, dyslipidemia, sex, smoking, and obesity.

Arterial remodeling was observed with advancing age in rhesus macaques and was characterized by an increased intimal thickness, higher matrix metalloproteinase 2 mRNA and protein expression, and enhanced angiotensin II signaling.220 Males tend to be at higher risk for CVD. In cynomolgus macaques, for example, aortic stiffness was more pronounced in males than in females, even though aortic pulse pressure similarly increased with aging in both males and females.173

Microarray analyses of the aorta indicated that, compared with females, aged males exhibited greater expression of genes associated with smooth muscle switching from a contractile to a secretory phenotype.173 Furthermore, while collagen density was maintained in both sexes of older cynomolgus monkeys, males displayed significant decreases in elastin density along with decreasing collagen type III and increasing collagen type VIII.172 In contrast, no sex differences in collagen isoform shifts were observed in aging rats, thereby promoting the use of nonhuman primates to examine sex-specific differences in aortic stiffness with aging.

Ex vivo studies further demonstrated changes associated with aging in endothelial cells derived from femoral arteries of nonhuman primates. Endothelial cells from older baboons exhibited a more rapid decline in endothelium-derived nitric oxide synthase levels ex vivo as a biomarker for vascular aging.188 Endothelial cells harvested from baboons fed a high-fat/high-sucrose diet demonstrated replicative senescence and increased expression of senescence-associated β-galactosidase independent of telomere length, and this model is being used further to study mechanisms of premature vascular senescence.189 Fewer endothelial colony-forming cells could be harvested from blood of aged versus younger rhesus macaques, and these cells were less able to form vessels after transplantation into immunodeficient mice.187 Ex vivo studies using vascular smooth muscle cells from rhesus macaques demonstrated that with increasing age, the nuclear factor erythroid–derived 2 pathway that regulates antioxidant responses was blunted or became dysfunctional, thereby inducing an increase in oxidative stress leading to NFκB activation and vascular inflammation.210 Studies in baboons indicated that clinical biomarkers of CVD were affected by genes that regulate lipid and lipoprotein metabolism.51 Heritable traits affecting risk for CVD unique to primates were examined in baboons and reported to be associated with obesity and red blood cell sodium-lithium countertransport activity, an indicator for salt-sensitive hypertension.51

Intervention strategies to inhibit the rate of vascular senescence are being tested in nonhuman primates. Human endothelial cells incubated ex vivo with serum from rhesus macaques undergoing CR exhibited increased angiogenesis and greater expression of vascular endothelial growth factors than cells incubated with serum from macaques fed ad libitum.54 Primary vascular smooth muscle cells from older rhesus macaques secreted higher levels of proinflammatory cytokines ex vivo (eg, IL-1β, MCP-1, TNF-α, IL-6) than did cells from younger animals, and this secretory profile was reversed by exposure to resveratrol, an anti-inflammatory and antiaging polyphenol.55 Resveratrol also inhibited the rate of arterial wall inflammation and stiffening in rhesus macaques fed a high-fat/high-sucrose diet.144

Nonhuman primates are being used to test hormonal intervention strategies related to risk for CVD. Estrogen deficiency, for example, is known to accelerate atherosclerosis during and after menopause. Estrogen replacement helps reduce development of coronary artery disease, but concerns exist about endometrial hyperplasia in the absence of combination hormone therapy with progesterone. As a result, studies in humans are being hampered by decreasing compliance due to a fear of breast cancer.153 In rhesus and cynomolgus macaques, however, an orally active progestin, nomegestrol acetate, was reported to counteract the endometrial stimulation of estrogen, and this could prove beneficial for continued development of hormone replacement therapy to ameliorate development of atherosclerosis in postmenopausal women.161 In addition, female cynomolgus monkeys fed a diet rich in soy protein and isoflavones exhibited a lower risk for CVD than monkeys fed a casein-lactalbumin diet.6

Musculoskeletal System

The overall degeneration of bones, joints, and muscles during aging contributes to frailty increasingly observed with advancing age. Bones undergo thinning through a loss of minerals (eg, calcium) and a shift from bone production by osteoblasts toward resorption by osteoclasts leading to osteoporosis. The accumulated wear and tear on cartilage and joints leads to calcification and mineral deposition producing less flexibility. Osteoporosis commonly progresses during aging via systemic loss of bone mass and deterioration of trabecular architecture that increases the risk for bone fractures.180 Skeletal muscle undergoes atrophy during aging from reduced production and loss of muscle fibers, reduced size of fibers, accumulation of mitochondrial DNA mutations, and deficiencies in cytochrome oxidase production that either alone or in combination affect strength and energy metabolism of the muscle.72 Chronic inflammation associated with aging contributes to the overall degeneration of the musculoskeletal system and further promotes osteoarthritis that is considered the most common cause of chronic disability in the elderly.135

Nonhuman primates, especially OWMs (eg, macaques, baboons), are particularly useful for studying changes in the musculoskeletal system because they develop bone and muscle loss similar to humans during aging.37,64,65,175 OWMs and nonhuman hominids (eg, great apes) display haversian osteonal remodeling of cortical bone that occurs in humans but not in rodents, and they have a similar reproductive endocrine system that affects bone metabolism.26,51,95,108

Increasing parity of up to 7 births by free-ranging rhesus macaque females is beneficial to retain bone mineral density and partially offsets the effects of bone loss from aging, while lower numbers of births are associated with earlier osteoporosis.35 While less frequently studied, baboons also display similar skeletal biology as humans with respect to bone composition and causes of fracture.51,96 Approximately 25% of older female baboons exhibit osteopenia and a steady decline in bone mineral density beginning at about 17 years of age. Unlike rhesus macaques, however, parity or interbirth intervals do not distinguish baboons with low versus higher bone marrow densities.96

Estrogen decline after menopause accelerates bone loss and decreasing bone mineral density in OWMs.20 In addition, ovariectomy in macaques reportedly mimics early and premenopausal changes that also occur in humans, such as remodeling and bone loss.31,108 The ovariectomy model has been especially important for evaluating effects of estrogen replacement therapy, bisphosphonates, odanacatib (a cathepsin K inhibitor), intraosseous recombinant human bone morphogenetic protein 2 / calcium phosphate matrix, and parathyroid hormone on progression of osteoporosis, osteopenia, and osteoarthritis. Results from several of these studies were consistent with those from human clinical trials.

Castration (orchidectomy) has been applied as a model for osteopenia/osteoporosis in male macaques and produced accelerated thinning of the skull, higher risk for vertebral fractures, kyphosis of the spine, and remodeling of vertebrae and femurs.118 In male rhesus macaques, vitamin K deficiency— considered a risk for bone loss in conjunction with the long-term use of anticoagulants—did not adversely affect skeletal changes if the animals were also administered sufficient vitamin D and calcium.25 Among NWMs, older marmoset females treated with alendronate, a bisphosphonate used to reduce bone loss, demonstrated improved trabecular volume and number, supporting their use to study human bone physiology during aging.17

Nonhuman primates are being used to address treatments for degenerative joint disease characterized by osteoarthritis and synovial inflammation (Suppl. Fig. 7). Acupuncture138 or treatment with glucosamine, chrondroitin, and polysulfated glycosaminoglycan along with corticosteroids and analgesics215 was reported to improve mobility in captive chimpanzees. Surgical removal of the femoral head and neck in a rhesus macaque with naturally occurring osteoarthritis also improved clinical outcome.67

Advancing age furthermore produces sarcopenia and reduces overall function in humans and nonhuman primates.48 Multiple mitochondrial DNA mutations increase in skeletal muscle cells of aging rhesus macaques similar to that in humans.130 In aging African green monkeys, there are changes in the myosin heavy chain isoforms, decreasing cross-sectional area of type I and type II fibers, and atrophy of type IIA fibers in the vastus lateralis that are also observed in aging humans.73

Reproductive System

Studies in captive and free-ranging nonhuman primates have examined reproductive senescence and postreproductive life span in relation to the time of interbirth intervals, declining rates of sexual activity, lack of observed breeding behaviors, and changes in social relationships and hormone levels in various species of nonhuman primates.11,13 The results suggest that there exists a regression in age-related reproductive decline whereby prosimians (eg, mouse lemurs) and some NWMs (tamarins) exhibit an abrupt reproductive decline near the end of life,199,225 while OWMs (eg, macaques, colobines, mangabeys) and some of the apes (eg, orangutans) display gradual but heterogeneous rates of reproductive senescence analogous to perimenopause,30,102,110,192 and chimps and gorillas undergo operational if not actual menopause.12,214 Histologically, ovaries of an aged captive sooty manabey, for example, exhibited a loss in distinct primordial and secondary follicles (Suppl. Fig. 8). Differences in reproductive decline may exist between wild and captive colonies of nonhuman primates. For example, orangutans in the wild continue to reproduce throughout life but in captivity display a postreproductive life span, despite wild and captive orangutans sharing similar maximum life spans.192 These findings suggest that animal management practices may affect reproduction patterns. Female NWMs such as marmosets and tamarins in wild and captive colonies do not exhibit gradual declines in fertility with aging, but infant mortality increases with the age of the mother, suggestive of declining maternal fitness with increasing age.179,199,225

Comparative analyses imply an evolutionary continuum. Extended postreproductive life span was not evident in the prosimians and callitrichid primates but developed in the cercopithecine or OWMs and some apes, followed by the occurrence of menopause in gorillas and humans that exhibit a long post-reproductive life span.11 Thus, menopause may have evolved to reduce reproduction as a means to conserve energy consumption and thus sustain female survival for care of offspring. Interestingly, human postreproductive life span evolved from increased longevity and reduced death rates without an increased reproductive life span. This distinction has been further studied in wild colonies of a prosimian species, 2 NWMs species, 2 OWM species and 2 great apes, in relation to a hunter-forager Dobe ! Kung population that experiences natural fertility and mortality resembling our preagricultural ancestors.3 Comparison of results in the 90th percentile of age at last live birth versus age at death showed that humans were unique and behaved as statistically significant outliers by exhibiting longer postreproductive life spans than nonhuman primates, which did fall along the correlative continuum. These findings thus argue in favor of an evolutionary role of human mothers and survival of their offspring via rationing of energy expense to support child-rearing over reproduction.

Nonhuman primate models of reproductive aging relate rates of pre-, peri-, and postmenopausal events to risks for developing chronic diseases of aging, such as coronary heart disease, osteoporosis, and cognitive decline.7,191 Disruptions in ovarian function are attributed to stress, anxiety, and depression in women and nonhuman primates, providing models to test effects of environment on health benefits associated with reproductive aging and risk for chronic diseases.115 Far less is known about reproductive senescence in males using nonhuman primate models, although declines in reproductive capacity were reported in male spider monkeys and rhesus macaques.98,182 Results from one study on male rhesus macaques suggested that reproductive decline was highly variable and related to social and demographic factors rather than aging body condition.22 In another study, supplemental administration of the androgens dehydroepiandrosterone and 5α-dihydrotestosterone to aged rhesus macaque males resulted in hormonal levels and circadian patterns similar to those in younger males, thereby supporting development of hormone therapies for treating elderly men.194 While aspects of reproductive senescence are found to be uniquely human, nonhuman primates continue to be relevant for testing intervention and treatment strategies that affect the postreproductive quality of life of humans.20

Genetics and Epigenetics

Aging appears to be somewhat delayed in humans, and comparative genetics studies that focused on humans and chimpanzees in relation to other organisms were performed to explore an evolutionary basis.62 The DNA genome sequences of humans and chimpanzees differ by 4%, and most of these differences reflect insertions and deletions; yet, aging begins later or is extended further in humans than in chimpanzees.49 From comparative genetics studies, de Magalhães and colleagues projected that aging did not necessarily result from programmatic sequential changes in gene expression or epigenetics, but they also found that the genetic changes observed between species in relation to aging were not entirely the result of random chance.61 They further suggested that multiple genetic regulatory processes established early in life and important during development are less subjected to selective pressure after reproduction and may become detrimental later in life. Thus, they hypothesized that natural selection pressures that occurred after humans and chimpanzees diverged mainly affected pathways for development and growth that subsequently also affected the longer life span in humans.8,62,75

Epigenetics is the study of changes in gene expression arising from changes in the genome that do not involve alterations in the DNA sequence, may be heritable, and are affected by aging. These include classic epigenetic mechanisms such as DNA methylation, histone modifications, and the more recently discovered noncoding RNAs (summarized in Table 2).59,228 Distinct, as well as global, changes in the epigenomic landscape occur during aging and cause aberrant gene expression contributing to a wide range of pathologic conditions, such as cancer, diabetes, obesity, and cardiovascular and neurodegenerative diseases.59,228

Table 2.

Epigenetic Changes and Aging.

Description of Epigenetic Change Studies in Nonhuman Primates
DNA methylation

Extensively studied epigenetic modification; involves the covalent addition of a
 methyl group to cytosines that are part of cytosine-guanine dinucleotides
 (CpG). Methylation of cytosines on both DNA strands results in full
 methylation. CpG dinucleotides generally occupy ~1% of the genome but are
 densely populated in specific regions of gene promoters that overlap with
 transcription start sites.59,228
  • None


Histone modifications

Histone N-terminal amino acid tails undergo a range of complex posttranslational
 modifications. These include acetylation, methylation, ubiquitination, and
 phosphorylation, which in turn recruit different coactivators or corepressors
 resulting in relaxed (transcriptional activation) or condensed chromatin
 (transcriptional repression).59,86,207,228
  • Global increase in H3K4me2 and H3K4me3 transcriptional activation in aging brains93

  • Elevated expression of H3K4me2 methyltransferases, namely SETD7 and DPY3093


RNA interference

miRNAs are single-stranded small noncoding RNAs that are ~20–22 nucleotides
 in length and highly conserved across species. Together with members of the
 Argonaut (Ago) family, they form the miRNA interference-silencing complex
 (miRISC). The miRISC regulates gene expression by binding to 3′ UTRs of
 mRNAs. Recent studies show that mRNA degradation may be the predominant
 mechanism of gene regulation by miRNAs.90
  • miR-34a mediated suppression of SIRT1 in intestines of SIV-infected rhesus macaques155

  • miR-142-3p mediated suppression of SIRT1 in brain macrophages of SIV-infected rhesus macaques38

Aging is associated with genome-wide alterations in the distribution of 5-methylcytosine in such a way that DNA methylation levels are decreased globally.113 While DNA methylation analysis has not been performed in nonhuman primates, increased gene expression of glucocorticoid receptor coincides with aging in rhesus macaques.27 The use of peripheral blood leukocytes is less invasive and experimentally convenient, but this approach has limitations, as it does not accurately reflect epigenetic changes occurring in the target tissues in vivo. The availability of the complete rhesus macaque genomic sequence; the physiologic, anatomic, and immunologic similarities between rhesus macaques and humans; and the feasibility for repetitive and longitudinal sampling from multiple tissues offer unparalleled advantages for using rhesus macaques in characterizing aging-related epigenetic changes affecting glucocorticoid receptor gene expression and for future testing of novel glucocorticoid receptor mimetics.

Histone modifications represent another well-studied epigenetic change that involves remodeling at the N-terminal group of lysine residues by various modifications such as acetylation, deacetylation, methylation, ADP-ribosylation, ubiquitination, and the like.59,86,207,228 Among these, histone acetylation, deacetylation, and methylation are considered the most predominant and best characterized posttranslational modifications. In rhesus macaque brains, aging is associated with a global increase in H3K4me2 and H3K4me3 transcriptional activation, suggesting the existence of open and active chromatin at transcription start sites and enhancer regions in the genome.93 In addition, H3K4me2 methyltransferases, namely SETD7 and DPY30, show elevated expression.93 The finding that H3K4me3 levels and the expression of its methyltransferases increase in yeast in response to double-stranded DNA damage response suggests a direct role for DNA damage response in triggering these specific modifications in brains of aged rhesus macaques.71 Performing brain and other vital/specialized organ-focused epigenetic studies in humans is very challenging, and the findings from this study highlight the feasibility and profound contributions that nonhuman primate models can make toward understanding aging-associated epigenetic changes in specific organs/tissues of interest over time.71

More recently, noncoding RNAs and microRNAs (miRNAs) in particular have been demonstrated to regulate the expression of genes linked to the aging process and include miR-34abc, miR-155, miR-21, miR-146a, miR-106ab, and miR-29ac.94 Although studies linking specific miRNAs to the aging process in rhesus macaques have yet to be performed, we and others have identified specific miRNAs that negatively regulate the anti-inflammatory and antiaging SIRT1 expression in the intestine155 and brain38 of chronic SIV-infected rhesus macaques. Increased miR-142-3p expression in brain macrophages resulted in decreased SIRT1 expression leading to macrophage activation and encephalitis.38 Similarly, we identified the miR-34a-SIRT1-acetylp65 axis to be a potential mediator of immune activation and persistent inflammation, also known as “inflamm-aging,” in the intestine.155

Conclusion

Nonhuman primates continue to provide valuable information for elucidating aging processes that also occur in humans, and their use in biomedical and psychosocial studies is expected to further contribute to improvements in healthy aging. While the majority of studies related to gerontology utilize rhesus and cynomolgus macaques, improvements in animal husbandry practices will likely expand the use of smaller nonhuman primates. With the growing development of antiaging strategies, it is expected that nonhuman primates will additionally be highly relevant for preclinical studies testing antiaging strategies.

Supplementary Material

Figure Legends
Supplemental Figures 1-4
Supplemental Figures 5-8

Acknowledgments

Funding

The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: Funding support was from the National Institutes of Health (OD01104 to all authors; R01DK083929 to M.M.; AI097059, AI110163, HL125054, and AI116198 to M.J.K.), and the Tulane Research Enhancement Fund (M.J.K. and E.S.D.).

Footnotes

Supplemental material for this article is available on the Veterinary Pathology website at http://vet.sagepub.com/supplemental.

*

References 11, 15, 16, 83, 99, 124, 126, 136, 165

References 23, 29, 77, 111, 126, 127, 166, 208

References 69, 82, 123, 133, 146, 176, 177

§

References 4, 36, 53, 68, 81, 157, 191, 199, 217–219

References 20, 47, 107, 108, 122, 162, 185, 198

Declaration of Conflicting Interests

The authors declare that they have no conflicts of interest with respect to their authorship or publication of this article.

References

  • 1.Aberg JA. Aging, inflammation, and HIV infection. Top Antivir Med. 2012;20(3):101–105. [PMC free article] [PubMed] [Google Scholar]
  • 2.Ailshire JA, Crimmins EM. Psychosocial factors associated with longevity in the United States: age differences between the old and oldest-old in the health and retirement study. J Aging Res. 2011;2011:530534. doi: 10.4061/2011/530534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Alberts SC, Altmann J, Brockman DK, et al. Reproductive aging patterns in primates reveal that humans are distinct. Proc Natl Acad Sci U S A. 2013;110(33):13440–13445. doi: 10.1073/pnas.1311857110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Almeida DV, Santos RR, Scalercio SR, et al. Morphological and morphometrical characterization, and estimation of population of preantral ovarian follicles from senile common squirrel monkey (Saimiri sciureus) Anim Reprod Sci. 2012;134(3–4):4–210. doi: 10.1016/j.anireprosci.2012.08.028. [DOI] [PubMed] [Google Scholar]
  • 5.Anderson KM, Wolf KN. Iron deficiency anemia in a ring-tailed lemur (Lemur catta) with concurrent chronic renal failure. J Am Vet Med Assoc. 2014;244(4):471–475. doi: 10.2460/javma.244.4.471. [DOI] [PubMed] [Google Scholar]
  • 6.Appt SE, Chen H, Goode AK, et al. The effect of diet and cardiovascular risk on ovarian aging in cynomolgus monkeys (Macaca fascicularis) Menopause (New York, NY) 2010;17(4):741–748. doi: 10.1097/gme.0b013e3181d20cd2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Appt SE, Ethun KF. Reproductive aging and risk for chronic disease: Insights from studies of nonhuman primates. Maturitas. 2010;67(1):7–14. doi: 10.1016/j.maturitas.2010.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Arbiza L, Dopazo J, Dopazo H. Positive selection, relaxation, and acceleration in the evolution of the human and chimp genome. PLoS Comput Biol. 2006;2(4):38. doi: 10.1371/journal.pcbi.0020038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Aspinall R, Pido-Lopez J, Imami N, et al. Old rhesus macaques treated with interleukin-7 show increased TREC levels and respond well to influenza vaccination. Rejuvenation Res. 2007;10(1):5–17. doi: 10.1089/rej.2006.9098. [DOI] [PubMed] [Google Scholar]
  • 10.Asquith M, Haberthur K, Brown M, et al. Age-dependent changes in innate immune phenotype and function in rhesus macaques (Macaca mulatta) Pathobiol Aging Age Relat Dis. 2012;2 doi: 10.3402/pba.v2i0.18052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Atsalis S, Margulis S. Primate reproductive aging: from lemurs to humans. Interdiscip Top Gerontol. 2008;36:186–194. doi: 10.1159/000137710. [DOI] [PubMed] [Google Scholar]
  • 12.Atsalis S, Videan E. Reproductive aging in captive and wild common chimpanzees: factors influencing the rate of follicular depletion. Am J Primatol. 2009;71(4):271–282. doi: 10.1002/ajp.20650. [DOI] [PubMed] [Google Scholar]
  • 13.Austad SN. Animal models of reproductive aging: what can they tell us? Ann N Y Acad Sci. 2010;1204:123–126. doi: 10.1111/j.1749-6632.2010.05609.x. [DOI] [PubMed] [Google Scholar]
  • 14.Austad SN. Comparative aging and life histories in mammals. Exp Gerontol. 1997;32(1–2):2–23. doi: 10.1016/s0531-5565(96)00059-9. [DOI] [PubMed] [Google Scholar]
  • 15.Austad SN. Comparative biology of aging. J Gerontol A Biol Sci Med Sci. 2009;64(2):199–201. doi: 10.1093/gerona/gln060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Austad SN, Fischer KE. The development of small primate models for aging research. ILAR J. 2011;52(1):78–88. doi: 10.1093/ilar.52.1.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bagi CM, Volberg M, Moalli M, et al. Age-related changes in marmoset trabecular and cortical bone and response to alendronate therapy resemble human bone physiology and architecture. Anat Rec (Hoboken) 2007;290(8):1005–1016. doi: 10.1002/ar.20561. [DOI] [PubMed] [Google Scholar]
  • 18.Bailey ME, Wang AC, Hao J, et al. Interactive effects of age and estrogen on cortical neurons: implications for cognitive aging. Neuroscience. 2011;191:148–158. doi: 10.1016/j.neuroscience.2011.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Barcia C, Ros CM, Ros-Bernal F, et al. Persistent phagocytic characteristics of microglia in the substantia nigra of long-term Parkinsonian macaques. J Neuroimmunol. 2013;261(1–2):2–60. doi: 10.1016/j.jneuroim.2013.05.001. [DOI] [PubMed] [Google Scholar]
  • 20.Bellino FL, Wise PM. Nonhuman primate models of menopause workshop. Biol Reprod. 2003;68(1):10–18. doi: 10.1095/biolreprod.102.005215. [DOI] [PubMed] [Google Scholar]
  • 21.Bendlin BB, Canu E, Willette A, et al. Effects of aging and calorie restriction on white matter in rhesus macaques. Neurobiol Aging. 2011;32(12):2319. doi: 10.1016/j.neurobiolaging.2010.04.008. e2311–e2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bercovitch F, Widdig A, Trefilov A, et al. A longitudinal study of age-specific reproductive output and body condition among male rhesus macaques, Macaca mulatta. Naturwissenschaften. 2003;90(7):309–312. doi: 10.1007/s00114-003-0436-1. [DOI] [PubMed] [Google Scholar]
  • 23.Bertrand A, Pasquier A, Petiet A, et al. Micro-MRI study of cerebral aging: ex vivo detection of hippocampal subfield reorganization, microhemorrhages and amyloid plaques in mouse lemur primates. PLoS One. 2013;8(2):e56593. doi: 10.1371/journal.pone.0056593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bihaqi SW, Zawia NH. Enhanced taupathy and AD-like pathology in aged primate brains decades after infantile exposure to lead (Pb) Neurotoxicology. 2013;39:95–101. doi: 10.1016/j.neuro.2013.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Binkley N, Krueger D, Engelke J, et al. Vitamin K deficiency from long-term warfarin anticoagulation does not alter skeletal status in male rhesus monkeys. J Bone Miner Res. 2007;22(5):695–700. doi: 10.1359/jbmr.070208. [DOI] [PubMed] [Google Scholar]
  • 26.Black A, Tilmont EM, Handy AM, et al. A nonhuman primate model of age-related bone loss: a longitudinal study in male and premenopausal female rhesus monkeys. Bone. 2001;28(3):295–302. doi: 10.1016/s8756-3282(00)00452-x. [DOI] [PubMed] [Google Scholar]
  • 27.Blalock EM, Grondin R, Chen KC, et al. Aging-related gene expression in hippocampus proper compared with dentate gyrus is selectively associated with metabolic syndrome variables in rhesus monkeys. J Neurosci. 2010;30(17):6058–6071. doi: 10.1523/JNEUROSCI.3956-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bodkin NL, Alexander TM, Ortmeyer HK, et al. Mortality and morbidity in laboratory-maintained rhesus monkeys and effects of long-term dietary restriction. J Gerontol A Biol Sci Med Sci. 2003;58(3):212–219. doi: 10.1093/gerona/58.3.b212. [DOI] [PubMed] [Google Scholar]
  • 29.Bons N, Silhol S, Barbie V, et al. A stereotaxic atlas of the grey lesser mouse lemur brain (Microcebus murinus) Brain Res Bull. 1998;46(1–2):2–1. doi: 10.1016/s0361-9230(97)00458-9. [DOI] [PubMed] [Google Scholar]
  • 30.Borries C, Koenig A. Reproductive and behavioral characteristics of aging in female Asian colobines. Interdiscip Top Gerontol. 2008;36:80–102. doi: 10.1159/000137686. [DOI] [PubMed] [Google Scholar]
  • 31.Brommage R. Perspectives on using nonhuman primates to understand the etiology and treatment of postmenopausal osteoporosis. J Musculoskelet Neuronal Interact. 2001;1(4):307–325. [PubMed] [Google Scholar]
  • 32.Bronikowski AM, Altmann J, Brockman DK, et al. Aging in the natural world: comparative data reveal similar mortality patterns across primates. Science. 2011;331(6022):1325–1328. doi: 10.1126/science.1201571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bukovsky A, Caudle MR, Svetlikova M, et al. Oogenesis in adult mammals, including humans: a review. Endocrine. 2005;26(3):301–316. doi: 10.1385/ENDO:26:3:301. [DOI] [PubMed] [Google Scholar]
  • 34.Carlyle BC, Nairn AC, Wang M, et al. cAMP-PKA phosphorylation of tau confers risk for degeneration in aging association cortex. Proc Natl Acad Sci U S A. 2014;111(13):5036–5041. doi: 10.1073/pnas.1322360111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cerroni AM, Tomlinson GA, Turnquist JE, et al. Effect of parity on bone mineral density in female rhesus macaques from Cayo Santiago. Am J Phys Anthropol. 2003;121(3):252–269. doi: 10.1002/ajpa.10238. [DOI] [PubMed] [Google Scholar]
  • 36.Chambers JK, Kuribayashi H, Ikeda S, et al. Distribution of neprilysin and deposit patterns of Abeta subtypes in the brains of aged squirrel monkeys (Saimiri sciureus) Amyloid. 2010;17(2):75–82. doi: 10.3109/13506129.2010.483119. [DOI] [PubMed] [Google Scholar]
  • 37.Champ JE, Binkley N, Havighurst T, et al. The effect of advancing age on bone mineral content of female rhesus monkeys. Bone. 1996;19(5):485–492. doi: 10.1016/s8756-3282(96)00243-8. [DOI] [PubMed] [Google Scholar]
  • 38.Chaudhuri AD, Yelamanchili SV, Marcondes MC, et al. Up-regulation of microRNA-142 in simian immunodeficiency virus encephalitis leads to repression of sirtuin1. FASEB J. 2013;27(9):3720–3729. doi: 10.1096/fj.13-232678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chen X, Errangi B, Li L, et al. Brain aging in humans, chimpanzees (Pan troglodytes), and rhesus macaques (Macaca mulatta): magnetic resonance imaging studies of macro- and microstructural changes. Neurobiol Aging. 2013;34(10):2248–2260. doi: 10.1016/j.neurobiolaging.2013.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Choi J, Li C, McDonald TJ, et al. Emergence of insulin resistance in juvenile baboon offspring of mothers exposed to moderate maternal nutrient reduction. Am J Physiol Regul Integr Comp Physiol. 2011;301(3):R757–R762. doi: 10.1152/ajpregu.00051.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cicin-Sain L, Messaoudi I, Park B, et al. Dramatic increase in naive T cell turn-over is linked to loss of naive T cells from old primates. Proc Natl Acad Sci U S A. 2007;104(50):19960–19965. doi: 10.1073/pnas.0705905104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Coe C, Lubach G, Kinnard J. Immune senescence in old and very old rhesus monkeys: reduced antibody response to influenza vaccination. Age (Dordr) 2012;34(5):1169–1177. doi: 10.1007/s11357-011-9356-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Coe CL. Biological and social predictors of immune senescence in the aged primate. Mech Ageing Dev. 2004;125(2):95–98. doi: 10.1016/j.mad.2003.11.011. [DOI] [PubMed] [Google Scholar]
  • 44.Coe CL, Ershler WB. Intrinsic and environmental influences on immune senescence in the aged monkey. Physiol Behav. 2001;73(3):379–384. doi: 10.1016/s0031-9384(01)00491-7. [DOI] [PubMed] [Google Scholar]
  • 45.Coe CL, Ershler WB, Champoux M, et al. Psychosocial factors and immune senescence in the aged primate. Ann N Y Acad Sci. 1992;650:276–282. doi: 10.1111/j.1749-6632.1992.tb49136.x. [DOI] [PubMed] [Google Scholar]
  • 46.Colman RJ, Anderson RM. Nonhuman primate calorie restriction. Antioxid Redox Signal. 2011;14(2):229–239. doi: 10.1089/ars.2010.3224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Colman RJ, Kemnitz JW, Lane MA, et al. Skeletal effects of aging and menopausal status in female rhesus macaques. J Clin Endocrinol Metab. 1999;84(11):4144–4148. doi: 10.1210/jcem.84.11.6151. [DOI] [PubMed] [Google Scholar]
  • 48.Colman RJ, McKiernan SH, Aiken JM, et al. Muscle mass loss in Rhesus monkeys: age of onset. Exp Gerontol. 2005;40(7):573–581. doi: 10.1016/j.exger.2005.05.001. [DOI] [PubMed] [Google Scholar]
  • 49.Consortium CSA. Initial sequence of the chimpanzee genome and comparison with the human genome. Nature. 2005;437(7055):69–87. doi: 10.1038/nature04072. [DOI] [PubMed] [Google Scholar]
  • 50.Costagliola D. Demographics of HIV and aging. Curr Opin HIV AIDS. 2014;9(4):294–301. doi: 10.1097/COH.0000000000000076. [DOI] [PubMed] [Google Scholar]
  • 51.Cox LA, Comuzzie AG, Havill LM, et al. Baboons as a model to study genetics and epigenetics of human disease. ILAR J. 2013;54(2):106–121. doi: 10.1093/ilar/ilt038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cruzen C, Colman RJ. Effects of caloric restriction on cardiovascular aging in non-human primates and humans. Clin Geriatr Med. 2009;25(4):733–743. doi: 10.1016/j.cger.2009.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Csiszar A, Podlutsky A, Podlutskaya N, et al. Testing the oxidative stress hypothesis of aging in primate fibroblasts: is there a correlation between species longevity and cellular ROS production? J Gerontol A Biol Sci Med Sci. 2012;67(8):841–852. doi: 10.1093/gerona/glr216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Csiszar A, Sosnowska D, Tucsek Z, et al. Circulating factors induced by caloric restriction in the nonhuman primate Macaca mulatta activate angiogenic processes in endothelial cells. J Gerontol A Biol Sci Med Sci. 2013;68(3):235–249. doi: 10.1093/gerona/gls158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Csiszar A, Sosnowska D, Wang M, et al. Age-associated proinflammatory secretory phenotype in vascular smooth muscle cells from the non-human primate Macaca mulatta: reversal by resveratrol treatment. J Gerontol A Biol Sci Med Sci. 2012;67(8):811–820. doi: 10.1093/gerona/glr228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Cuozzo FP, Sauther ML, Gould L, et al. Variation in dental wear and tooth loss among known-aged, older ring-tailed lemurs (Lemur catta): a comparison between wild and captive individuals. Am J Primatol. 2010;72(11):1026–1037. doi: 10.1002/ajp.20846. [DOI] [PubMed] [Google Scholar]
  • 57.Dal-Pan A, Pifferi F, Marchal J, et al. Cognitive performances are selectively enhanced during chronic caloric restriction or resveratrol supplementation in a primate. PLoS One. 2011;6(1):e16581. doi: 10.1371/journal.pone.0016581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dal-Pan A, Terrien J, Pifferi F, et al. Caloric restriction or resveratrol supplementation and ageing in a non-human primate: first-year outcome of the RESTRIKAL study in Microcebus murinus. Age (Dordr) 2011;33(1):15–31. doi: 10.1007/s11357-010-9156-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Daniel M, Tollefsbol T. Epigenetic linkage of aging, cancer and nutrition. J Exp Biol. 2015;218:59–70. doi: 10.1242/jeb.107110. pt 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Darusman HS, Pandelaki J, Mulyadi R, et al. Poor memory performance in aged cynomolgus monkeys with hippocampal atrophy, depletion of amyloid beta 1–42 and accumulation of tau proteins in cerebrospinal fluid. In Vivo. 2014;28(2):173–184. [PubMed] [Google Scholar]
  • 61.de Magalhães JP. Programmatic features of aging originating in development: aging mechanisms beyond molecular damage? FASEB J. 2012;26(12):4821–4826. doi: 10.1096/fj.12-210872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.de Magalhães JP, Church GM. Analyses of human-chimpanzee orthologous gene pairs to explore evolutionary hypotheses of aging. Mech Ageing Dev. 2007;128(5–6):6–355. doi: 10.1016/j.mad.2007.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.de Magalhães JP, Costa J, Church GM. An analysis of the relationship between metabolism, developmental schedules, and longevity using phylogenetic independent contrasts. J Gerontol A Biol Sci Med Sci. 2007;62(2):149–160. doi: 10.1093/gerona/62.2.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.DeRousseau C. Aging in the musculoskeletal system of rhesus monkeys: III. Bone loss. Am J Phys Anthropol. 1985;68(2):157–167. doi: 10.1002/ajpa.1330680203. [DOI] [PubMed] [Google Scholar]
  • 65.DeRousseau CJ, Rawlins RG, Denlinger JL. Aging in the musculoskeletal system of rhesus monkeys: I. Passive joint excursion. Am J Phys Anthropol. 1983;61(4):483–494. doi: 10.1002/ajpa.1330610411. [DOI] [PubMed] [Google Scholar]
  • 66.Didier ES, Sugimoto C, Bowers LC, et al. Immune correlates of aging in outdoor-housed captive rhesus macaques (Macaca mulatta) Immun Ageing. 2012;9(1):25. doi: 10.1186/1742-4933-9-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dufour JP, Phillippi-Falkenstein K, Bohm RP, et al. Excision of femoral head and neck for treatment of coxofemoral degenerative joint disease in a rhesus macaque (Macaca mulatta) Comp Med. 2012;62(6):539–542. [PMC free article] [PubMed] [Google Scholar]
  • 68.Elfenbein HA, Rosen RF, Stephens SL, et al. Cerebral beta-amyloid angiopathy in aged squirrel monkeys. Histol Histopathol. 2007;22(2):155–167. doi: 10.14670/HH-22.155. [DOI] [PubMed] [Google Scholar]
  • 69.Engelberth RC, Silva KD, Azevedo CV, et al. Morphological changes in the suprachiasmatic nucleus of aging female marmosets (Callithrix jacchus) Biomed Res Int. 2014;2014:243825. doi: 10.1155/2014/243825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Fan XT, Zhao F, Ai Y, et al. Cortical glutamate levels decrease in a non-human primate model of dopamine deficiency. Brain Res. 2014;1552:34–40. doi: 10.1016/j.brainres.2013.12.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Faucher D, Wellinger RJ. Methylated H3K4, a transcription-associated histone modification, is involved in the DNA damage response pathway. PLos Genetics. 2010;6(8):e1001082. doi: 10.1371/journal.pgen.1001082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Faulkner JA, Larkin LM, Claflin DR, et al. Age-related changes in the structure and function of skeletal muscles. Clin Exp Pharmacol Physiol. 2007;34(11):1091–1096. doi: 10.1111/j.1440-1681.2007.04752.x. [DOI] [PubMed] [Google Scholar]
  • 73.Feng X, Zhang T, Xu Z, et al. Myosin heavy chain isoform expression in the vastus lateralis muscle of aging African green vervet monkeys. Exp Gerontol. 2012;47(8):601–607. doi: 10.1016/j.exger.2012.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Fiandaca MS, Federoff HJ. Using viral-mediated gene delivery to model Parkinson’s disease: do nonhuman primate investigations expand our understanding? Exp Neurol. 2014;256:117–125. doi: 10.1016/j.expneurol.2013.03.014. [DOI] [PubMed] [Google Scholar]
  • 75.Finch CE. Evolution of the human lifespan and diseases of aging: roles of infection, inflammation, and nutrition. Proc Natl Acad Sci U S A. 2010;107(suppl 1):1718–1724. doi: 10.1073/pnas.0909606106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Finch CE, Austad SN. Commentary: is Alzheimer’s disease uniquely human? Neurobiol Aging. 2015;36(2):553–555. doi: 10.1016/j.neurobiolaging.2014.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Finch CE, Austad SN. Primate aging in the mammalian scheme: the puzzle of extreme variation in brain aging. Age (Dordr) 2012;34(5):1075–1091. doi: 10.1007/s11357-011-9355-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fiori JL, Shin YK, Kim W, et al. Resveratrol prevents beta-cell dedifferentiation in nonhuman primates given a high-fat/high-sugar diet. Diabetes. 2013;62(10):3500–3513. doi: 10.2337/db13-0266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Franceschi C. Inflammaging as a major characteristic of old people: can it be prevented or cured? Nutr Rev. 2007;65(12):S173–S176. doi: 10.1111/j.1753-4887.2007.tb00358.x. pt 2. [DOI] [PubMed] [Google Scholar]
  • 80.Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69(suppl 1):S4–S9. doi: 10.1093/gerona/glu057. [DOI] [PubMed] [Google Scholar]
  • 81.Garber PA, Leigh SR. Ontogenetic variation in small-bodied New World primates: implications for patterns of reproduction and infant care. Folia Primatol (Basel) 1997;68(1):1–22. doi: 10.1159/000157226. [DOI] [PubMed] [Google Scholar]
  • 82.Geula C, Nagykery N, Wu CK. Amyloid-beta deposits in the cerebral cortex of the aged common marmoset (Callithrix jacchus): incidence and chemical composition. Acta Neuropathol. 2002;103(1):48–58. doi: 10.1007/s004010100429. [DOI] [PubMed] [Google Scholar]
  • 83.Gomes NM, Ryder OA, Houck ML, et al. Comparative biology of mammalian telomeres: hypotheses on ancestral states and the roles of telomeres in longevity determination. Aging Cell. 2011;10(5):761–768. doi: 10.1111/j.1474-9726.2011.00718.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gomez D, Barbosa A, Thery M, et al. Age affects photoentrainment in a nocturnal primate. J Biol Rhythms. 2012;27(2):164–171. doi: 10.1177/0748730411435223. [DOI] [PubMed] [Google Scholar]
  • 85.Goncharova ND, Lapin BA. Age-related endocrine dysfunction in nonhuman primates. Ann N Y Acad Sci. 2004;1019:321–325. doi: 10.1196/annals.1297.054. [DOI] [PubMed] [Google Scholar]
  • 86.Goossens-Beumer IJ, Benard A, van Hoesel AQ, et al. Age-dependent clinical prognostic value of histone modifications in colorectal cancer. Transl Res. 2015;165(5):578–588. doi: 10.1016/j.trsl.2014.11.001. [DOI] [PubMed] [Google Scholar]
  • 87.Grabli D, Karachi C, Folgoas E, et al. Gait disorders in parkinsonian monkeys with pedunculopontine nucleus lesions: a tale of two systems. J Neurosci. 2013;33(29):11986–11993. doi: 10.1523/JNEUROSCI.1568-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gray DT, Engle JR, Recanzone GH. Age-related neurochemical changes in the rhesus macaque cochlear nucleus. J Comp Neurol. 2014;522(7):1527–1541. doi: 10.1002/cne.23479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gray DT, Engle JR, Rudolph ML, et al. Regional and age-related differences in GAD67 expression of parvalbumin- and calbindin-expressing neurons in the rhesus macaque auditory midbrain and brainstem. J Comp Neurol. 2014;522(18):4074–4084. doi: 10.1002/cne.23659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Guo H, Ingolia NT, Weissman JS, et al. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 2010;466(7308):835–840. doi: 10.1038/nature09267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Haberthur K, Engelman F, Barron A, et al. Immune senescence in aged nonhuman primates. Exp Gerontol. 2010;45(9):655–661. doi: 10.1016/j.exger.2010.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Haley GE, Kohama SG, Urbanski HF, et al. Age-related decreases in SYN levels associated with increases in MAP-2, apoE, and GFAP levels in the rhesus macaque prefrontal cortex and hippocampus. Age (Dordr) 2010;32(3):283–296. doi: 10.1007/s11357-010-9137-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Han Y, Han D, Yan Z, et al. Stress-associated H3K4 methylation accumulates during postnatal development and aging of rhesus macaque brain. Aging Cell. 2012;11(6):1055–1064. doi: 10.1111/acel.12007. [DOI] [PubMed] [Google Scholar]
  • 94.Harries LW. MicroRNAs as mediators of the ageing process. Genes (Basel) 2014;5(3):656–670. doi: 10.3390/genes5030656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Havill LM, Allen MR, Harris JA, et al. Intracortical bone remodeling variation shows strong genetic effects. Calcif Tissue Int. 2013;93(5):472–480. doi: 10.1007/s00223-013-9775-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Havill LM, Levine SM, Newman DE, et al. Osteopenia and osteoporosis in adult baboons (Papio hamadryas) J Med Primatol. 2008;37(3):146–153. doi: 10.1111/j.1600-0684.2007.00270.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Henderson M, Urbanc B, Cruz L. A computational model for the loss of neuronal organization in microcolumns. Biophys J. 2014;106(10):2233–2242. doi: 10.1016/j.bpj.2014.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Hernandez-Lopez L, Cerda-Molina AL, Diaz-Diaz G, et al. Aging-related reproductive decline in the male spider monkey (Ateles geoffroyi) J Med Primatol. 2012;41(2):115–121. doi: 10.1111/j.1600-0684.2011.00528.x. [DOI] [PubMed] [Google Scholar]
  • 99.Heuer E, Rosen RF, Cintron A, et al. Nonhuman primate models of Alzheimer-like cerebral proteopathy. Curr Pharm Des. 2012;18(8):1159–1169. doi: 10.2174/138161212799315885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.High KP, Brennan-Ing M, Clifford DB, et al. HIV and aging: state of knowledge and areas of critical need for research. A report to the NIH Office of AIDS Research by the HIV and Aging Working Group. J Acquir Immune Defic Syndr. 2012;60(suppl 1):S1–S18. doi: 10.1097/QAI.0b013e31825a3668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Hoffman CL, Higham JP, Heistermann M, et al. Immune function and HPA axis activity in free-ranging rhesus macaques. Physiol Behav. 2011;104(3):507–514. doi: 10.1016/j.physbeh.2011.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hoffman CL, Higham JP, Mas-Rivera A, et al. Terminal investment and senescence in rhesus macaques (Macaca mulatta) on Cayo Santiago. Behav Ecol. 2010;21(5):972–978. doi: 10.1093/beheco/arq098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ingram DK, Roth GS. Glycolytic inhibition as a strategy for developing calorie restriction mimetics. Exp Gerontol. 2011;46(2–3):3–148. doi: 10.1016/j.exger.2010.12.001. [DOI] [PubMed] [Google Scholar]
  • 104.Ingram DK, Roth GS, Lane MA, et al. The potential for dietary restriction to increase longevity in humans: extrapolation from monkey studies. Biogerontology. 2006;7(3):143–148. doi: 10.1007/s10522-006-9013-2. [DOI] [PubMed] [Google Scholar]
  • 105.Ingram DK, Zhu M, Mamczarz J, et al. Calorie restriction mimetics: an emerging research field. Aging Cell. 2006;5(2):97–108. doi: 10.1111/j.1474-9726.2006.00202.x. [DOI] [PubMed] [Google Scholar]
  • 106.Jankovic V, Messaoudi I, Nikolich-Zugich J. Phenotypic and functional T-cell aging in rhesus macaques (Macaca mulatta): differential behavior of CD4 and CD8 subsets. Blood. 2003;102(9):3244–3251. doi: 10.1182/blood-2003-03-0927. [DOI] [PubMed] [Google Scholar]
  • 107.Jerome CP. Hormonal therapies and osteoporosis. ILAR J. 2004;45(2):170–178. doi: 10.1093/ilar.45.2.170. [DOI] [PubMed] [Google Scholar]
  • 108.Jerome CP, Peterson PE. Nonhuman primate models in skeletal research. Bone. 2001;29(1):1–6. doi: 10.1016/s8756-3282(01)00477-x. [DOI] [PubMed] [Google Scholar]
  • 109.Jimenez-Gomez Y, Mattison JA, Pearson KJ, et al. Resveratrol improves adipose insulin signaling and reduces the inflammatory response in adipose tissue of rhesus monkeys on high-fat, high-sugar diet. Cell Metab. 2013;18(4):533–545. doi: 10.1016/j.cmet.2013.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Johnson RL, Kapsalis E. Heterogeneity of reproductive aging in free-ranging female rhesus macaques. Interdiscip Top Gerontol. 2008;36:62–79. doi: 10.1159/000137680. [DOI] [PubMed] [Google Scholar]
  • 111.Joly M, Ammersdorfer S, Schmidtke D, et al. Touchscreen-based cognitive tasks reveal age-related impairment in a primate aging model, the grey mouse lemur (Microcebus murinus) PLoS One. 2014;9(10):e109393. doi: 10.1371/journal.pone.0109393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Josset L, Engelmann F, Haberthur K, et al. Increased viral loads and exacerbated innate host responses in aged macaques infected with the 2009 pandemic H1N1 influenza A virus. J Virol. 2012;86(20):11115–11127. doi: 10.1128/JVI.01571-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Jung M, Pfeifer G. Aging and DNA methylation. BMC Biol. 2015;13(7):015–0118. doi: 10.1186/s12915-015-0118-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kanaan NM, Kordower JH, Collier TJ. Age and region-specific responses of microglia, but not astrocytes, suggest a role in selective vulnerability of dopamine neurons after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure in monkeys. Glia. 2008;56(11):1199–1214. doi: 10.1002/glia.20690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kaplan JR. Origins and health consequences of stress-induced ovarian dysfunction. Interdiscip Top Gerontol. 2008;36:162–185. doi: 10.1159/000137709. [DOI] [PubMed] [Google Scholar]
  • 116.Kastman EK, Willette AA, Coe CL, et al. A calorie-restricted diet decreases brain iron accumulation and preserves motor performance in old rhesus monkeys. J Neurosci. 2012;32(34):11897–11904. doi: 10.1523/JNEUROSCI.2553-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kemnitz JW. Calorie restriction and aging in nonhuman primates. ILAR J. 2011;52(1):66–77. doi: 10.1093/ilar.52.1.66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kessler MJ, Wang Q, Cerroni AM, et al. Long-term effects of castration on the skeleton of male rhesus monkeys (Macaca mulatta) Am J Primatol. doi: 10.1002/ajp.22399. [published online March 16, 2015] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kimura N, Okabayashi S, Ono F. Dynein dysfunction disrupts beta-amyloid clearance in astrocytes through endocytic disturbances. Neuroreport. 2014;25(7):514–520. doi: 10.1097/WNR.0000000000000124. [DOI] [PubMed] [Google Scholar]
  • 120.Kling HM, Shipley TW, Guyach S, et al. Trimethoprim-sulfamethoxazole treatment does not reverse obstructive pulmonary changes in pneumocystiscolonized nonhuman primates with SHIV infection. J Acquir Immune Defic Syndr. 2014;65(4):381–389. doi: 10.1097/QAI.0000000000000007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kroner Z. The relationship between Alzheimer’s disease and diabetes: type 3 diabetes? Altern Med Rev. 2009;14(4):373–379. [PubMed] [Google Scholar]
  • 122.Krueger D, Todd H, Haffa A, et al. Central region-of-interest analysis of lumbar spine densitometry demonstrates lower bone mass in older rhesus monkeys. Bone. 1999;24(1):29–33. doi: 10.1016/s8756-3282(98)00145-8. [DOI] [PubMed] [Google Scholar]
  • 123.Lacreuse A, Chang J, Metevier CM, et al. Oestradiol modulation of cognition in adult female marmosets (Callithrix jacchus) J Neuroendocrinol. 2014;26(5):296–309. doi: 10.1111/jne.12147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Lane MA. Nonhuman primate models in biogerontology. Exp Gerontol. 2000;35(5):533–541. doi: 10.1016/s0531-5565(00)00102-9. [DOI] [PubMed] [Google Scholar]
  • 125.Languille S, Aujard F, Pifferi F. Effect of dietary fish oil supplementation on the exploratory activity, emotional status and spatial memory of the aged mouse lemur, a non-human primate. Behav Brain Res. 2012;235(2):280–286. doi: 10.1016/j.bbr.2012.08.014. [DOI] [PubMed] [Google Scholar]
  • 126.Languille S, Blanc S, Blin O, et al. The grey mouse lemur: a non-human primate model for ageing studies. Ageing Res Rev. 2012;11(1):150–162. doi: 10.1016/j.arr.2011.07.001. [DOI] [PubMed] [Google Scholar]
  • 127.Languille S, Lievin-Bazin A, Picq JL, et al. Deficits of psychomotor and mnesic functions across aging in mouse lemur primates. Front Behav Neurosci. 2014;8:446. doi: 10.3389/fnbeh.2014.00446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Lankau EW, Turner PV, Mullan RJ, et al. Use of nonhuman primates in research in North America. J Am Assoc Lab Anim Sci. 2014;53(3):278–282. [PMC free article] [PubMed] [Google Scholar]
  • 129.Laurent S. Defining vascular aging and cardiovascular risk. J Hypertens. 2012;30:S3–S8. doi: 10.1097/HJH.0b013e328353e501. [DOI] [PubMed] [Google Scholar]
  • 130.Lee CM, Chung SS, Kaczkowski JM, et al. Multiple mitochondrial DNA deletions associated with age in skeletal muscle of rhesus monkeys. J Gerontol. 1993;48(6):B201–B205. doi: 10.1093/geronj/48.6.b201. [DOI] [PubMed] [Google Scholar]
  • 131.Lee SH, Min KJ. Caloric restriction and its mimetics. BMB Rep. 2013;46(4):181–187. doi: 10.5483/BMBRep.2013.46.4.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Lin ZY, Imamura M, Sano C, et al. Molecular signatures to define spermatogenic cells in common marmoset (Callithrix jacchus) Reproduction. 2012;143(5):597–609. doi: 10.1530/REP-11-0215. [DOI] [PubMed] [Google Scholar]
  • 133.Liu JV, Bock NA, Silva AC. Rapid high-resolution three-dimensional mapping of T1 and age-dependent variations in the non-human primate brain using magnetization-prepared rapid gradient-echo (MPRAGE) sequence. Neuroimage. 2011;56(3):1154–1163. doi: 10.1016/j.neuroimage.2011.02.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Liu Y, Yue F, Tang R, et al. Progressive loss of striatal dopamine terminals in MPTP-induced acute parkinsonism in cynomolgus monkeys using vesicular monoamine transporter type 2 PET imaging ([(18)F]AV-133) Neurosci Bull. 2014;30(3):409–416. doi: 10.1007/s12264-013-1374-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Loeser RF. Age-related changes in the musculoskeletal system and the development of osteoarthritis. Clin Geriatr Med. 2010;26(3):371–386. doi: 10.1016/j.cger.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lowenstine LJ. A primer of primate pathology: lesions and nonlesions. Toxicol Pathol. 2003;31(suppl):92–102. doi: 10.1080/01926230390177668. [DOI] [PubMed] [Google Scholar]
  • 137.Lowery EM, Brubaker AL, Kuhlmann E, et al. The aging lung. Clin Interv Aging. 2013;8:1489–1496. doi: 10.2147/CIA.S51152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Magden ER, Haller RL, Thiele EJ, et al. Acupuncture as an adjunct therapy for osteoarthritis in chimpanzees (Pan troglodytes) J Am Assoc Lab Anim Sci. 2013;52(4):475–480. [PMC free article] [PubMed] [Google Scholar]
  • 139.Makris N, Papadimitriou GM, van der Kouwe A, et al. Frontal connections and cognitive changes in normal aging rhesus monkeys: a DTI study. Neurobiol Aging. 2007;28(10):1556–1567. doi: 10.1016/j.neurobiolaging.2006.07.005. [DOI] [PubMed] [Google Scholar]
  • 140.Marchal J, Blanc S, Epelbaum J, et al. Effects of chronic calorie restriction or dietary resveratrol supplementation on insulin sensitivity markers in a primate, Microcebus murinus. PLoS One. 2012;7(3):e34289. doi: 10.1371/journal.pone.0034289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Marchal J, Dal-Pan A, Epelbaum J, et al. Calorie restriction and resveratrol supplementation prevent age-related DNA and RNA oxidative damage in a non-human primate. Exp Gerontol. 2013;48(9):992–1000. doi: 10.1016/j.exger.2013.07.002. [DOI] [PubMed] [Google Scholar]
  • 142.Marchal J, Dorieux O, Haro L, et al. Characterization of blood biochemical markers during aging in the Grey Mouse Lemur (Microcebus murinus): impact of gender and season. BMC Vet Res. 2012;8:211. doi: 10.1186/1746-6148-8-211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Mattison JA, Roth GS, Beasley TM, et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature. 2012;489(7415):318–321. doi: 10.1038/nature11432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Mattison JA, Wang M, Bernier M, et al. Resveratrol prevents high fat/sucrose diet-induced central arterial wall inflammation and stiffening in nonhuman primates. Cell Metab. 2014;20(1):183–190. doi: 10.1016/j.cmet.2014.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.McCay C, Crowell M, Maynard L. The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. Nutrition. 1989;5(3):155–171. [PubMed] [Google Scholar]
  • 146.McCormack AL, Mak SK, Di Monte DA. Increased alpha-synuclein phosphorylation and nitration in the aging primate substantia nigra. Cell Death Dis. 2012;3:e315. doi: 10.1038/cddis.2012.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.McDonald Pavelka MS. The nonhuman primate perspective: old age, kinship and social partners in a monkey society. J Cross Cult Gerontol. 1994;9(2):219–229. doi: 10.1007/BF00972151. [DOI] [PubMed] [Google Scholar]
  • 148.Mendelsohn AR, Larrick JW. Dietary restriction: critical co-factors to separate health span from life span benefits. Rejuvenation Res. 2012;15(5):523–529. doi: 10.1089/rej.2012.1371. [DOI] [PubMed] [Google Scholar]
  • 149.Messaoudi I, Estep R, Robinson B, et al. Nonhuman primate models of human immunology. Antioxid Redox Signal. 2011;14(2):261–273. doi: 10.1089/ars.2010.3241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Messaoudi I, Fischer M, Warner J, et al. Optimal window of caloric restriction onset limits its beneficial impact on T-cell senescence in primates. Aging Cell. 2008;7(6):908–919. doi: 10.1111/j.1474-9726.2008.00440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Messaoudi I, Warner J, Fischer M, et al. Delay of T cell senescence by caloric restriction in aged long-lived nonhuman primates. Proc Natl Acad Sci U S A. 2006;103(51):19448–19453. doi: 10.1073/pnas.0606661103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Meyer C, Kerns A, Haberthur K, et al. Improving immunity in the elderly: current and future lessons from nonhuman primate models. Age (Dordr) 2012;34(5):1157–1168. doi: 10.1007/s11357-011-9353-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Mikkola TS, Clarkson TB. Estrogen replacement therapy, atherosclerosis, and vascular function. Cardiovasc Res. 2002;53(3):605–619. doi: 10.1016/s0008-6363(01)00466-7. [DOI] [PubMed] [Google Scholar]
  • 154.Minor RK, Allard JS, Younts CM, et al. Dietary interventions to extend life span and health span based on calorie restriction. J Gerontol A Biol Sci Med Sci. 2010;65(7):695–703. doi: 10.1093/gerona/glq042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Mohan M, Kumar V, Lackner AA, et al. Dysregulated miR-34a-SIRT1-acetyl p65 axis is a potential mediator of immune activation in the colon during chronic simian immunodeficiency virus infection of rhesus macaques. J Immunol. 2015;194(1):291–306. doi: 10.4049/jimmunol.1401447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Morin N, Jourdain VA, Morissette M, et al. Long-term treatment with l-DOPA and an mGlu5 receptor antagonist prevents changes in brain basal ganglia dopamine receptors, their associated signaling proteins and neuropeptides in parkinsonian monkeys. Neuropharmacology. 2014;79:688–706. doi: 10.1016/j.neuropharm.2014.01.014. [DOI] [PubMed] [Google Scholar]
  • 157.Nievergelt CM, Martin RD. Energy intake during reproduction in captive common marmosets (Callithrix jacchus) Physiol Behav. 1999;65(4–5):5–849. doi: 10.1016/s0031-9384(98)00249-2. [DOI] [PubMed] [Google Scholar]
  • 158.Nilsson PM, Boutouyrie P, Laurent S. Vascular aging: a tale of EVA and ADAM in cardiovascular risk assessment and prevention. Hypertension. 2009;54(1):3–10. doi: 10.1161/HYPERTENSIONAHA.109.129114. [DOI] [PubMed] [Google Scholar]
  • 159.Nyirenda MJ, Carter R, Tang JI, et al. Prenatal programming of metabolic syndrome in the common marmoset is associated with increased expression of 11beta-hydroxysteroid dehydrogenase type 1. Diabetes. 2009;58(12):2873–2879. doi: 10.2337/db09-0873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Pandrea I, Landay A, Wilson C, et al. Using the pathogenic and nonpathogenic nonhuman primate model for studying non-AIDS comorbidities. Curr HIV/ AIDS Rep. 2015;12(1):54–67. doi: 10.1007/s11904-014-0245-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Paris JM, Williams KJ, Hermsmeyer KR, et al. Nomegestrol acetate and vascular reactivity: nonhuman primate experiments. Steroids. 2000;65(10–11):621–627. doi: 10.1016/s0039-128x(00)00118-5. [DOI] [PubMed] [Google Scholar]
  • 162.Pennypacker B, Chen C, Zheng H, et al. Inhibition of cathepsin K increases modeling-based bone formation, and improves cortical dimension and strength in adult ovariectomized monkeys. J Bone Miner Res. 2014;29(8):1847–1858. doi: 10.1002/jbmr.2211. [DOI] [PubMed] [Google Scholar]
  • 163.Peters A. Structural changes in the normally aging cerebral cortex of primates. Prog Brain Res. 2002;136:455–465. doi: 10.1016/s0079-6123(02)36038-2. [DOI] [PubMed] [Google Scholar]
  • 164.Peters A, Sethares C. Aging and the myelinated fibers in prefrontal cortex and corpus callosum of the monkey. J Comp Neurol. 2002;442(3):277–291. doi: 10.1002/cne.10099. [DOI] [PubMed] [Google Scholar]
  • 165.Phillips KA, Sherwood CC. Age-related differences in corpus callosum area of capuchin monkeys. Neuroscience. 2012;202:202–208. doi: 10.1016/j.neuroscience.2011.11.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Picq JL. Aging affects executive functions and memory in mouse lemur primates. Exp Gerontol. 2007;42(3):223–232. doi: 10.1016/j.exger.2006.09.013. [DOI] [PubMed] [Google Scholar]
  • 167.Picq JL, Aujard F, Volk A, et al. Age-related cerebral atrophy in nonhuman primates predicts cognitive impairments. Neurobiol Aging. 2012;33(6):1096–1109. doi: 10.1016/j.neurobiolaging.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Pifferi F, Rahman A, Languille S, et al. Effects of dietary resveratrol on the sleep-wake cycle in the non-human primate gray mouse lemur (Microcebus murinus) Chronobiol Int. 2012;29(3):261–270. doi: 10.3109/07420528.2011.654019. [DOI] [PubMed] [Google Scholar]
  • 169.Pifl C, Rajput A, Reither H, et al. Is Parkinson’s disease a vesicular dopamine storage disorder? Evidence from a study in isolated synaptic vesicles of human and nonhuman primate striatum. J Neurosci. 2014;34(24):8210–8218. doi: 10.1523/JNEUROSCI.5456-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Polverino F, Doyle-Eisele M, McDonald J, et al. A novel nonhuman primate model of cigarette smoke-induced airway disease. Am J Pathol. 2015;185(3):741–755. doi: 10.1016/j.ajpath.2014.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Potts LF, Wu H, Singh A, et al. Modeling Parkinson’s disease in monkeys for translational studies, a critical analysis. Exp Neurol. 2014;256:133–143. doi: 10.1016/j.expneurol.2013.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Qiu H, Depre C, Ghosh K, et al. Mechanism of gender-specific differences in aortic stiffness with aging in nonhuman primates. Circulation. 2007;116(6):669–676. doi: 10.1161/CIRCULATIONAHA.107.689208. [DOI] [PubMed] [Google Scholar]
  • 173.Qiu H, Tian B, Resuello RG, et al. Sex-specific regulation of gene expression in the aging monkey aorta. Physiol Genomics. 2007;29(2):169–180. doi: 10.1152/physiolgenomics.00229.2006. [DOI] [PubMed] [Google Scholar]
  • 174.Rahman A, Languille S, Lamberty Y, et al. Sleep deprivation impairs spatial retrieval but not spatial learning in the non-human primate grey mouse lemur. PLoS One. 2013;8(5):e64493. doi: 10.1371/journal.pone.0064493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Reinwald S, Burr D. Review of nonprimate, large animal models for osteoporosis research. J Bone Miner Res. 2008;23(9):1353–1368. doi: 10.1359/JBMR.080516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Ridley RM, Baker HF, Windle CP, et al. Very long term studies of the seeding of beta-amyloidosis in primates. J Neural Transm. 2006;113(9):1243–1251. doi: 10.1007/s00702-005-0385-2. [DOI] [PubMed] [Google Scholar]
  • 177.Risser L, Dolius L, Fonta C, et al. Diffeomorphic registration with selfadaptive spatial regularization for the segmentation of non-human primate brains. Conf Proc IEEE Eng Med Biol Soc. 2014;2014:6695–6698. doi: 10.1109/EMBC.2014.6945164. [DOI] [PubMed] [Google Scholar]
  • 178.Ross CN, Power ML, Artavia JM, et al. Relation of food intake behaviors and obesity development in young common marmoset monkeys. Obesity (Silver Spring) 2013;21(9):1891–1899. doi: 10.1002/oby.20432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Rutherford JN, deMartelly VA, Layne Colon DG, et al. Developmental origins of pregnancy loss in the adult female common marmoset monkey (Callithrix jacchus) PLoS One. 2014;9(5):e96845. doi: 10.1371/journal.pone.0096845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Ryan TM, Shaw CN. Gracility of the modern Homo sapiens skeleton is the result of decreased biomechanical loading. Proc Natl Acad Sci U S A. 2015;112(2):372–377. doi: 10.1073/pnas.1418646112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Sandell JH, Peters A. Disrupted myelin and axon loss in the anterior commissure of the aged rhesus monkey. J Comp Neurol. 2003;466(1):14–30. doi: 10.1002/cne.10859. [DOI] [PubMed] [Google Scholar]
  • 182.Schlatt S, Pohl CR, Ehmcke J, et al. Age-related changes in diurnal rhythms and levels of gonadotropins, testosterone, and inhibin B in male rhesus monkeys (Macaca mulatta) Biol Reprod. 2008;79(1):93–99. doi: 10.1095/biolreprod.107.066126. [DOI] [PubMed] [Google Scholar]
  • 183.Schmucker D, Owen R, Outenreath R, et al. Basis for the age-related decline in intestinal mucosal immunity. Clin Dev Immunol. 2003;10(2–4):4–167. doi: 10.1080/10446670310001642168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Schultz C, Dick EJ, Cox AB, et al. Expression of stress proteins alpha B-crystallin, ubiquitin, and hsp27 in pallido-nigral spheroids of aged rhesus monkeys. Neurobiol Aging. 2001;22(4):677–682. doi: 10.1016/s0197-4580(01)00229-9. [DOI] [PubMed] [Google Scholar]
  • 185.Seeherman H, Li X, Smith E, et al. Intraosseous injection of rhBMP-2/calcium phosphate matrix improves bone structure and strength in the proximal aspect of the femur in chronic ovariectomized nonhuman primates. J Bone Joint Surg Am. 2013;95(1):36–47. doi: 10.2106/JBJS.K.00668. [DOI] [PubMed] [Google Scholar]
  • 186.Shaykhiev R, Crystal RG. Innate immunity and chronic obstructive pulmonary disease: a mini-review. Gerontology. 2013;59(6):481–489. doi: 10.1159/000354173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Shelley WC, Leapley AC, Huang L, et al. Changes in the frequency and in vivo vessel-forming ability of rhesus monkey circulating endothelial colonyforming cells across the lifespan (birth to aged) Pediatr Res. 2012;71(2):156–161. doi: 10.1038/pr.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Shi Q, Aida K, Vandeberg JL, et al. Passage-dependent changes in baboon endothelial cells: relevance to in vitro aging. DNA Cell Biol. 2004;23(8):502–509. doi: 10.1089/1044549041562294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Shi Q, Hubbard GB, Kushwaha RS, et al. Endothelial senescence after highcholesterol, high-fat diet challenge in baboons. Am J Physiol Heart Circ Physiol. 2007;292(6):H2913–H2920. doi: 10.1152/ajpheart.01405.2006. [DOI] [PubMed] [Google Scholar]
  • 190.Shipley TW, Kling HM, Morris A, et al. Persistent pneumocystis colonization leads to the development of chronic obstructive pulmonary disease in a nonhuman primate model of AIDS. J Infect Dis. 2010;202(2):302–312. doi: 10.1086/653485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Shively CA, Clarkson TB. The unique value of primate models in translational research: nonhuman primate models of women’s health: introduction and overview. Am J Primatol. 2009;71(9):715–721. doi: 10.1002/ajp.20720. [DOI] [PubMed] [Google Scholar]
  • 192.Shumaker RW, Wich SA, Perkins L. Reproductive life history traits of female orangutans (Pongo spp) Interdiscip Top Gerontol. 2008;36:147–161. doi: 10.1159/000137705. [DOI] [PubMed] [Google Scholar]
  • 193.Smith DL, Jr, Nagy TR, Allison DB. Calorie restriction: what recent results suggest for the future of ageing research. Eur J Clin Invest. 2010;40(5):440–450. doi: 10.1111/j.1365-2362.2010.02276.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Sorwell KG, Kohama SG, Urbanski HF. Testosterone increases circulating dehydroepiandrosterone sulfate levels in the male rhesus macaque. Front Endocrinol (Lausanne) 2014;5:101. doi: 10.3389/fendo.2014.00101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Sridharan A, Bendlin BB, Gallagher CL, et al. Effect of age and calorie restriction on corpus callosal integrity in rhesus macaques: a fiber tractography study. Neurosci Lett. 2014;569:38–42. doi: 10.1016/j.neulet.2014.03.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Sridharan A, Pehar M, Salamat MS, et al. Calorie restriction attenuates astrogliosis but not amyloid plaque load in aged rhesus macaques: a preliminary quantitative imaging study. Brain Res. 2013;1508:1–8. doi: 10.1016/j.brainres.2013.02.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Steinert S, White DM, Zou Y, et al. Telomere biology and cellular aging in nonhuman primate cells. Exp Cell Res. 2002;272(2):146–152. doi: 10.1006/excr.2001.5409. [DOI] [PubMed] [Google Scholar]
  • 198.Tanko LB, Karsdal MA, Christiansen C. The clinical potential of estrogen for the prevention of osteoarthritis: what is known and what needs to be done? Womens Health (Lond Engl) 2005;1(1):125–132. doi: 10.2217/17455057.1.1.125. [DOI] [PubMed] [Google Scholar]
  • 199.Tardif SD, Araujo A, Arruda MF, et al. Reproduction and aging in marmosets and tamarins. Interdiscip Top Gerontol. 2008;36:29–48. doi: 10.1159/000137678. [DOI] [PubMed] [Google Scholar]
  • 200.Tardif SD, Power ML, Ross CN, et al. Characterization of obese phenotypes in a small nonhuman primate, the common marmoset (Callithrix jacchus) Obesity (Silver Spring) 2009;17(8):1499–1505. doi: 10.1038/oby.2009.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Taylor L, Daniels C, Schmucker D. Ageing compromises gastrointestinal mucosal immune response in the rhesus monkey. Immunology. 1992;75(4):614–618. [PMC free article] [PubMed] [Google Scholar]
  • 202.Terrien J, Ambid L, Nibbelink M, et al. Non-shivering thermogenesis activation and maintenance in the aging gray mouse lemur (Microcebus murinus) Exp Gerontol. 2010;45(6):442–448. doi: 10.1016/j.exger.2010.03.013. [DOI] [PubMed] [Google Scholar]
  • 203.Terrien J, Blanc S, Zizzari P, et al. Physiological responses to chronic heat exposure in an aging non-human primate species, the gray mouse lemur (Microcebus murinus) Exp Gerontol. 2011;46(9):747–754. doi: 10.1016/j.exger.2011.05.007. [DOI] [PubMed] [Google Scholar]
  • 204.Terrien J, Perret M, Aujard F. Gender markedly modulates behavioral thermoregulation in a non-human primate species, the mouse lemur (Microcebus murinus) Physiol Behav. 2010;101(4):469–473. doi: 10.1016/j.physbeh.2010.07.012. [DOI] [PubMed] [Google Scholar]
  • 205.Terrien J, Zizzari P, Bluet-Pajot MT, et al. Effects of age on thermoregulatory responses during cold exposure in a nonhuman primate, Microcebus murinus. Am J Physiol Regul Integr Comp Physiol. 2008;295(2):R696–R703. doi: 10.1152/ajpregu.00629.2007. [DOI] [PubMed] [Google Scholar]
  • 206.Tigno XT, Gerzanich G, Hansen BC. Age-related changes in metabolic parameters of nonhuman primates. J Gerontol A Biol Sci Med Sci. 2004;59(11):1081–1088. doi: 10.1093/gerona/59.11.1081. [DOI] [PubMed] [Google Scholar]
  • 207.Tollefsbol TO. Dietary epigenetics in cancer and aging. Cancer Treat Res. 2014;159:257–267. doi: 10.1007/978-3-642-38007-5_15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Trouche SG, Maurice T, Rouland S, et al. The three-panel runway maze adapted to Microcebus murinus reveals age-related differences in memory and perseverance performances. Neurobiol Learn Mem. 2010;94(1):100–106. doi: 10.1016/j.nlm.2010.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Tsukada H, Nishiyama S, Ohba H, et al. Comparing amyloid-beta deposition, neuroinflammation, glucose metabolism, and mitochondrial complex I activity in brain: a PET study in aged monkeys. Eur J Nucl Med Mol Imaging. 2014;41(11):2127–2136. doi: 10.1007/s00259-014-2821-8. [DOI] [PubMed] [Google Scholar]
  • 210.Ungvari Z, Bailey-Downs L, Gautam T, et al. Age-associated vascular oxidative stress, Nrf2 dysfunction, and NF-fkappagB activation in the nonhuman primate Macaca mulatta. J Gerontol A Biol Sci Med Sci. 2011;66(8):866–875. doi: 10.1093/gerona/glr092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Uno H. Age-related pathology and biosenescent markers in captive rhesus macaques. Age (Omaha) 1997;20(1):1–13. doi: 10.1007/s11357-997-0001-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Urbanski HF, Sorwell KG. Age-related changes in neuroendocrine rhythmic function in the rhesus macaque. Age (Dordr) 2012;34(5):1111–1121. doi: 10.1007/s11357-011-9352-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Vaccari M, Franchini G. Memory T cells in Rhesus macaques. Adv Exp Med Biol. 2010;684:126–144. doi: 10.1007/978-1-4419-6451-9_10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Videan EN, Fritz J, Heward CB, et al. Reproductive aging in female chimpanzees (Pan troglodytes) Interdiscip Top Gerontol. 2008;36:103–118. doi: 10.1159/000137688. [DOI] [PubMed] [Google Scholar]
  • 215.Videan EN, Lammey ML, Lee DR. Diagnosis and treatment of degenerative joint disease in a captive male chimpanzee (Pan troglodytes) J Am Assoc Lab Anim Sci. 2011;50(2):263–266. [PMC free article] [PubMed] [Google Scholar]
  • 216.Voytko ML, Tinkler GP. Cognitive function and its neural mechanisms in nonhuman primate models of aging, Alzheimer disease, and menopause. Front Biosci. 2004;9:1899–1914. doi: 10.2741/1370. [DOI] [PubMed] [Google Scholar]
  • 217.Walker LC, Kitt CA, Schwam E, et al. Senile plaques in aged squirrel monkeys. Neurobiol Aging. 1987;8(4):291–296. doi: 10.1016/0197-4580(87)90067-4. [DOI] [PubMed] [Google Scholar]
  • 218.Walker LC, Masters C, Beyreuther K, et al. Amyloid in the brains of aged squirrel monkeys. Acta Neuropathol. 1990;80(4):381–387. doi: 10.1007/BF00307691. [DOI] [PubMed] [Google Scholar]
  • 219.Walker ML, Anderson DC, Herndon JG, et al. Ovarian aging in squirrel monkeys (Saimiri sciureus) Reproduction. 2009;138(5):793–799. doi: 10.1530/REP-08-0449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Wang M, Takagi G, Asai K, et al. Aging increases aortic MMP-2 activity and angiotensin II in nonhuman primates. Hypertension. 2003;41(6):1308–1316. doi: 10.1161/01.HYP.0000073843.56046.45. [DOI] [PubMed] [Google Scholar]
  • 221.Willette AA, Bendlin BB, McLaren DG, et al. Age-related changes in neural volume and microstructure associated with interleukin-6 are ameliorated by a calorie-restricted diet in old rhesus monkeys. Neuroimage. 2010;51(3):987–994. doi: 10.1016/j.neuroimage.2010.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Willette AA, Coe CL, Colman RJ, et al. Calorie restriction reduces psychological stress reactivity and its association with brain volume and microstructure in aged rhesus monkeys. Psychoneuroendocrinology. 2012;37(7):903–916. doi: 10.1016/j.psyneuen.2011.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Willette AA, Gallagher C, Bendlin BB, et al. Homocysteine, neural atrophy, and the effect of caloric restriction in rhesus monkeys. Neurobiol Aging. 2012;33(4):670–680. doi: 10.1016/j.neurobiolaging.2010.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Wisconsin National Primate Research Center Library Primate taxonomy. http://pin.primate.wisc.edu/aboutp/taxonomy/. Published 2008.
  • 225.Wright P, King SJ, Baden A, et al. Aging in wild female lemurs: sustained fertility with increased infant mortality. Interdiscip Top Gerontol. 2008;36:17–28. doi: 10.1159/000137677. [DOI] [PubMed] [Google Scholar]
  • 226.Wu D, Yue F, Zou C, et al. Analysis of glucose metabolism in cynomolgus monkeys during aging. Biogerontology. 2012;13(2):147–155. doi: 10.1007/s10522-011-9364-1. [DOI] [PubMed] [Google Scholar]
  • 227.Yue F, Lu C, Ai Y, et al. Age-associated changes of cerebrospinal fluid amyloid-beta and tau in cynomolgus monkeys. Neurobiol Aging. 2014;35(7):1656–1659. doi: 10.1016/j.neurobiolaging.2014.01.139. [DOI] [PubMed] [Google Scholar]
  • 228.Zane L, Sharma V, Misteli T. Common features of chromatin in aging and cancer: cause or coincidence? Trends Cell Biol. 2014;24(11):686–694. doi: 10.1016/j.tcb.2014.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Zhao G, Guo S, Somel M, et al. Evolution of human longevity uncoupled from caloric restriction mechanisms. PLoS One. 2014;9(1):e84117. doi: 10.1371/journal.pone.0084117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Zhou Y, Sun M, Li H, et al. Recovery of behavioral symptoms in hemiparkinsonian rhesus monkeys through combined gene and stem cell therapy. Cytotherapy. 2013;15(4):467–480. doi: 10.1016/j.jcyt.2013.01.007. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Figure Legends
Supplemental Figures 1-4
Supplemental Figures 5-8

RESOURCES