1
|
Metzler LAP, Metzger JM, Gerred KJ, Emborg ME, Kapoor A. Expression patterns of blood-based biomarkers of neurodegeneration and inflammation across adulthood in rhesus macaques. Exp Gerontol 2025; 203:112736. [PMID: 40122475 PMCID: PMC11998483 DOI: 10.1016/j.exger.2025.112736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
As the global human population rapidly ages and diseases of aging become more prevalent, preclinical models of age-related neurodegenerative disorders are increasingly important for identifying early diagnostic biomarkers, monitoring disease progression, and evaluating treatment responsiveness. Rhesus macaques are an ideal species for studies on neurodegeneration due to their phylogenetic relatedness to humans and their complex brain anatomy and physiology. Technological advances in assay sensitivity have facilitated the identification of blood-based biomarkers of neurodegeneration and inflammation in human populations. The aim of this study was to translate these methods for use in male and female rhesus macaques across adulthood. We collected plasma samples from 47 rhesus macaques representing pre-adult (1-5 years, n = 6 female, n = 5 male), young (5-7 years, n = 5 female, n = 7 male), middle (8-16 years, n = 7 female, n = 7 male), and older adult (17-22 years, n = 6 female, n = 4 male) subjects. Quantified biomarkers included neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), amyloid beta (Aβ42, Aβ40, and their ratio), total tau, phosphorylated tau (pTau181), interleukin (IL) 2, IL-6, IL-8, and IL-10. Plasma NfL and IL-6 levels were significantly increased with age in both sexes, with a marked rise during middle adulthood. The ratio of Aβ42/Aβ40 was significantly lower in the middle and older aged females compared to the youngest group. There was no effect of age or sex on total tau or pTau181 levels. Overall, these results demonstrate the feasibility of evaluating blood biomarkers of neurodegeneration and inflammation in rhesus macaques during adulthood.
Collapse
Affiliation(s)
- Ludwig A P Metzler
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Jeanette M Metzger
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Keenan J Gerred
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Marina E Emborg
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA; Department of Medical Physics, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Amita Kapoor
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| |
Collapse
|
2
|
Jeanpierre GM, Rausch MK, Santacruz SR. Mechanical properties of fresh rhesus monkey brain tissue. Acta Biomater 2025; 196:233-243. [PMID: 40015355 DOI: 10.1016/j.actbio.2025.02.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Studying brain tissue mechanics is critical for understanding how the brain's physical properties influence its biological functions. Non-human primates, such as rhesus monkeys, are a key translational model for human neuroscience research, yet their brain tissue mechanics remain poorly understood. We report the mechanical properties of rhesus monkey white (corona radiata, CR) and gray (basal ganglia, BG) matter during compression relaxation, tension relaxation, tension-compression cycling (strain = 0.15, nCR = 21, nBG = 14), and shear cycling (strain = 0.3, nCR = 17, nBG = 9). Compression relaxation yields short and long-term time constants of 1.13 ± 0.041 s and 26.3 ± 0.68 s for CR and 1.22 ± 0.046 s and 28.3 ± 0.70 s for BG. Tension relaxation yields short and long-term time constants of 1.10 ± 0.052 s and 28.2 ± 0.82 s for CR and 1.19 ± 0.052 s and 29 ± 1.3 s for BG. Tension-compression cycling yields elastic moduli (E₁, E₂, E₃) of 36 ± 3.8 kPa, 0.61 ± 0.096 kPa, and 9.3 ± 0.90 kPa for CR and 27 ± 4.8 kPa, 0.68 ± 0.092 kPa, and 8 ± 1.0 kPa for BG. Shear cycling yields E₁, E₂, and E₃ of 3.9 ± 0.77 kPa, 0.19 ± 0.034 kPa, and 3.1 ± 0.40 kPa for CR and 2.8 ± 0.52 kPa, 0.18 ± 0.058 kPa, and 3.2 ± 0.53 kPa for BG. Hysteresis areas are also captured during tension-compression and shear cycling. These findings extend the translatability of rhesus monkey models for neuroscience. STATEMENT OF SIGNIFICANCE: While rhesus monkeys are a valuable translational model in human neuroscience research, there is a huge gap in knowledge about rhesus monkey brain tissue mechanics. This study serves to increase our understanding of rhesus monkey brain tissue mechanics and is the first to report the stiffness, time constant, and hysteresis parameters for rhesus monkey brain tissue in compression, tension, and shear for both the corona radiata and basal ganglia. The data is available in an open-source format, allowing others to fit and validate their mechanical models.
Collapse
Affiliation(s)
- Grace M Jeanpierre
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Manuel K Rausch
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA; Department of Aerospace Engineering & Engineering Mechanics, The University of Texas at Austin, Austin, TX, USA; Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Samantha R Santacruz
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Interdisciplinary Neuroscience Program, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
3
|
Lin J, Yu J, Wang X, Shi R, Liang Y, Li J, Zhou T, Chen C, Duan X, Deng Y, Yang S, Zeng S, Shen X, Chen X, Wang Y, Sun G, Shu Z. Research progress on the anti-aging effect of polysaccharides of traditional Chinese medicine: Using Caenorhabditis elegans as an animal model. FASEB J 2025; 39:e70454. [PMID: 40085128 DOI: 10.1096/fj.202403250rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
With the growing elderly population and increasing incidence of various aging-related diseases, the scientific community is faced with an urgent challenge to identify natural anti-aging agents. Traditional Chinese medicine (TCM) polysaccharides have been proven to have good anti-aging activities. This article reviews the literature on the anti-aging pathways of traditional Chinese medicine polysaccharides applied to Caenorhabditis elegans models in the past decade. In our study, we found that 45 TCM polysaccharides from 28 genera and 26 families could delay the aging process of C. elegans. Traditional Chinese medicine polysaccharides delay the aging of C. elegans mainly by anti-oxidative stress, eliminating free radicals, repairing DNA damage, and insulin/insulin-like growth factor signaling pathway (IIS signaling pathway). In addition, an increasing number of traditional Chinese medicine polysaccharides have been found to prolong the lifespan of C. elegans by reducing inflammation, regulating intestinal flora, and affecting immune cell function. In this paper, C. elegans was used as an animal model to clarify the anti-aging pathway of traditional Chinese medicine polysaccharides, so as to provide theoretical guidance for future research and clinical experiments on the anti-aging effect of traditional Chinese medicine polysaccharides.
Collapse
Affiliation(s)
- Jiazi Lin
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Jiamin Yu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Xiao Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Ruixiang Shi
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Yefang Liang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Jianhua Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Tong Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Chengkai Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Xiaodong Duan
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Yongan Deng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Simin Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Shuting Zeng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Xuejuan Shen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Xiangyu Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Yi Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guibo Sun
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zunpeng Shu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| |
Collapse
|
4
|
Vanderlip CR, Jutras ML, Asch PA, Zhu SY, Lerma MN, Buffalo EA, Glavis-Bloom C. Parallel patterns of age-related working memory impairment in marmosets and macaques. Aging (Albany NY) 2025; 17:778-797. [PMID: 40131878 PMCID: PMC11984434 DOI: 10.18632/aging.206225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
As humans age, some experience cognitive impairment while others do not. When impairment does occur, it is not expressed uniformly across cognitive domains and varies in severity across individuals. Translationally relevant model systems are critical for understanding the neurobiological drivers of this variability, which is essential to uncovering the mechanisms underlying the brain's susceptibility to the effects of aging. As such, non-human primates (NHPs) are particularly important due to shared behavioral, neuroanatomical, and age-related neuropathological features with humans. For many decades, macaque monkeys have served as the primary NHP model for studying the neurobiology of cognitive aging. More recently, the common marmoset has emerged as an advantageous model for this work due to its short lifespan that facilitates longitudinal studies. Despite their growing popularity as a model, whether marmosets exhibit patterns of age-related cognitive impairment comparable to those observed in macaques and humans remains unexplored. To address this major limitation for the development and evaluation of the marmoset as a model of cognitive aging, we directly compared working memory ability as a function of age in macaques and marmosets on the identical task. We also implemented varying delays to further tax working memory capacity. Our findings demonstrate that marmosets and macaques exhibit remarkably similar age-related working memory deficits, with macaques performing better than marmosets on longer delays. These results highlight the similarities and differences between the two most commonly used NHP models and support the value of the marmoset as a model for cognitive aging research within the neuroscience community.
Collapse
Affiliation(s)
- Casey R. Vanderlip
- Systems Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Megan L. Jutras
- Department of Neurobiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Payton A. Asch
- Systems Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Stephanie Y. Zhu
- Department of Neurobiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Monica N. Lerma
- Department of Neurobiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth A. Buffalo
- Department of Neurobiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Courtney Glavis-Bloom
- Systems Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Hass JK, Fernandes AG, Montague MJ, Burgos-Rodriguez A, Martinez MI, Brent LJN, Snyder-Mackler N, Danias J, Wollstein G, Higham JP, Melin AD. The ocular surface microbiome of rhesus macaques. RESEARCH SQUARE 2025:rs.3.rs-6205866. [PMID: 40162216 PMCID: PMC11952637 DOI: 10.21203/rs.3.rs-6205866/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background The ocular surface microbiome (OSM) is important for eye health, and variations in OSM composition have been associated with multiple diseases in humans. Studies of OSM-disease dynamics in humans are confounded by lifestyle factors. Animal models provide a complementary approach to understanding biological systems, free from many confounds of human studies. Here, we provide the first study of the OSM of rhesus macaques, a premier animal model for eye health and disease. We describe the taxonomy of the rhesus macaque OSM, and explore compositional correlations with age, sex, and living condition. Methods We analyzed eyelid and conjunctival microbiota swabs from 132 individual rhesus macaques (Macaca mulatta) (57 males, 75 females, 1-26 years old) from one captive and one free-ranging group using 16S V3/V4 MiSeq sequencing. We investigated alpha diversity, beta diversity, and differential abundance. Results We found several similarities between the top Phyla and Genera of the rhesus macaque OSM and those reported in human literature. Significantly higher alpha diversity, which may reflect age-related ocular surface mucous membrane integrity and immune function, was present in younger individuals compared to older ones. Higher alpha diversity was also present in free-ranging rhesus macaques compared to ones in captivity, possibly related to differences in diet, exercise, and medical exposures between macaques in different living conditions. Beta diversity was most strongly influenced by individual identity, followed by living conditions. Sex did not correlate with any OSM variation. Conclusions In this study we describe the taxonomic composition of the rhesus macaque OSM, and identify significant differences in alpha and beta diversity according to individual nonhuman primate host variables and the surrounding environment. Our findings suggest composition of the nonhuman primate OSM is shaped by age-related physiology, individual identity, and external living conditions. Our results offer novel insights into an underexplored region of the primate microbiome and highlight the utility of rhesus macaques as a model system for investigating the links between the OSM, ocular health, and disease.
Collapse
Affiliation(s)
- Joelle K Hass
- University of Calgary (Department of Anthropology and Archaeology)
| | | | | | | | | | | | | | - John Danias
- SUNY Downstate Health Sciences University (Department of Ophthalmology)
| | | | - James P Higham
- New York University College of Arts & Science (Department of Anthropology)
| | - Amanda D Melin
- University of Calgary (Department of Anthropology and Archaeology)
| |
Collapse
|
6
|
Cui Z, Dong Y, Sholl J, Lu J, Raubenheimer D. The Rhesus Macaque as an Animal Model for Human Nutrition: An Ecological-Evolutionary Perspective. Annu Rev Anim Biosci 2025; 13:441-464. [PMID: 39556489 DOI: 10.1146/annurev-animal-111523-102354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Nutrition is a complex and contested area in biomedicine, which requires diverse evidence sources. Nonhuman primate models are considered an important biomedical research tool because of their biological similarities to humans, but they are typically used with little explicit consideration of their ecology and evolution. Using the rhesus macaque (RM), we consider the potential of nutritional ecology for enriching the use of primates as models for human nutrition. We introduce some relevant aspects of RM evolutionary and social ecology and discuss two examples where they have been used in biomedical research: obesity and aging. We next consider how insights from nutritional ecology can help inform and direct the use of RM as a biomedical model. We conclude by illustrating how conceptual tools might inform the use of RM as a model for human nutrition and extracting insights from RM that might be relevant to broader theoretical considerations around animal model systems.
Collapse
Affiliation(s)
- Zhenwei Cui
- Institute of Biodiversity and Ecology, Zhengzhou University, Zhengzhou, Henan, China
- Centre for Nutritional Ecology, Centre for Sport Nutrition and Health, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, Henan, China
| | - Yunlong Dong
- Institute of Biodiversity and Ecology, Zhengzhou University, Zhengzhou, Henan, China
- Centre for Nutritional Ecology, Centre for Sport Nutrition and Health, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, Henan, China
| | - Jonathan Sholl
- ImmunoConcept Lab, Université de Bordeaux, Collège Sciences de la Santé, CNRS UMR 5164, Bordeaux, France
| | - Jiqi Lu
- Institute of Biodiversity and Ecology, Zhengzhou University, Zhengzhou, Henan, China
| | - David Raubenheimer
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia;
- Centre for Nutritional Ecology, Centre for Sport Nutrition and Health, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Huber HF, Ainsworth HC, Quillen EE, Salmon A, Ross C, Azhar AD, Bales K, Basso MA, Coleman K, Colman R, Darusman HS, Hopkins W, Hotchkiss CE, Jorgensen MJ, Kavanagh K, Li C, Mattison JA, Nathanielsz PW, Saputro S, Scorpio DG, Sosa PM, Vallender EJ, Wang Y, Zeiss CJ, Shively CA, Cox LA. Comparative lifespan and healthspan of nonhuman primate species common to biomedical research. GeroScience 2025; 47:135-151. [PMID: 39585646 PMCID: PMC11872815 DOI: 10.1007/s11357-024-01421-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
There is a critical need to generate age- and sex-specific survival curves to characterize chronological aging consistently across nonhuman primates (NHP) used in biomedical research. Sex-specific Kaplan-Meier survival curves were computed in 12 translational aging models: baboon, bonnet macaque, chimpanzee, common marmoset, coppery titi monkey, cotton-top tamarin, cynomolgus macaque, Japanese macaque, pigtail macaque, rhesus macaque, squirrel monkey, and vervet/African green. After employing strict inclusion criteria, primary results are based on 12,269 NHPs that survived to adulthood and died of natural/health-related causes. A secondary analysis was completed for 32,616 NHPs that died of any cause. Results show a pattern of reduced male survival among catarrhines (African and Asian primates), especially macaques, but not platyrrhines (Central and South American primates). For many species, median lifespans were lower than previously reported. An important consideration is that these analyses may offer a better reflection of healthspan than lifespan since research NHPs are typically euthanized for humane welfare reasons before their natural end of life. This resource represents the most comprehensive characterization of sex-specific lifespan and age-at-death distributions for 12 biomedically relevant species, to date. These results clarify relationships among NHP ages and provide a valuable resource for the aging research community, improving human-NHP age equivalencies, informing investigators of expected survival rates, providing a metric for comparisons in future studies, and contributing to understanding of factors driving lifespan differences within and among species.
Collapse
Affiliation(s)
| | | | - Ellen E Quillen
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Adam Salmon
- University of Texas Health Science Center, San Antonio, TX, USA
| | - Corinna Ross
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Adinda D Azhar
- Primate Research Center, IPB University, Bogor, Indonesia
| | - Karen Bales
- California National Primate Research Center, Davis, CA, USA
- University of California, Davis, CA, USA
| | - Michele A Basso
- Washington National Primate Research Center, Seattle, WA, USA
| | - Kristine Coleman
- Oregon National Primate Research Center, Hillsboro, OR, USA
- Oregon Health & Science University, Portland, OR, USA
| | - Ricki Colman
- Wisconsin National Primate Research Center, Madison, WI, USA
| | - Huda S Darusman
- Primate Research Center, IPB University, Bogor, Indonesia
- School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - William Hopkins
- The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | | | | | - Kylie Kavanagh
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
- University of Tasmania, Hobart, TAS, Australia
| | - Cun Li
- University of Wyoming, Laramie, WY, USA
| | - Julie A Mattison
- National Institute On Aging, National Institutes of Health, Gaithersburg, MD, USA
| | - Peter W Nathanielsz
- Texas Biomedical Research Institute, San Antonio, TX, USA
- University of Wyoming, Laramie, WY, USA
| | - Suryo Saputro
- Primate Research Center, IPB University, Bogor, Indonesia
| | - Diana G Scorpio
- Texas Biomedical Research Institute, San Antonio, TX, USA
- Envol Biomedical, Immokalee, FL, USA
| | | | - Eric J Vallender
- Tulane National Primate Research Center, Covington, LA, USA
- New England Primate Research Center, Southborough, MA, USA
| | - Yaomin Wang
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Carol A Shively
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Laura A Cox
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
8
|
Wang Q, Francis G. Coming to the Caribbean: Eighty-five years of rhesus macaques (Macaca mulatta) at Cayo Santiago-A rare nonhuman primate model for the studies of adaptation, diseases, genetics, natural disasters, and resilience. Am J Primatol 2025; 87:e23659. [PMID: 38961812 DOI: 10.1002/ajp.23659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/16/2024] [Indexed: 07/05/2024]
Abstract
The Cayo Santiago rhesus macaque colony represents one of the most important nonhuman primate resources since their introduction to the Caribbean area in 1938. The 85 years of continuing existence along with the comprehensive database of the rhesus colony and the derived skeletal collections have provided and will continue to provide a powerful tool to test hypotheses about adaptive and evolutionary mechanisms in both biology and medicine.
Collapse
Affiliation(s)
- Qian Wang
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Texas, USA
| | - George Francis
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Texas, USA
| |
Collapse
|
9
|
Cooper EB, Whalen C, Beeby N, Negron-Del Valle JE, Phillips D, Snyder-Mackler N, Brent LJN, Higham JP. Associations between social behaviour and proinflammatory immune activation are modulated by age in a free-ranging primate population. Anim Behav 2025; 219:123021. [PMID: 39829684 PMCID: PMC11741183 DOI: 10.1016/j.anbehav.2024.10.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The effect of the social environment on the proinflammatory immune response may mediate the relationship between social environment and fitness but remains understudied outside captive animals and human populations. Age can also influence both immune function and social behaviour, and hence may modulate their relationships. This study investigates the role of social interactions in driving the concentrations of two urinary markers of proinflammatory immune activation, neopterin and soluble urokinase plasminogen activator receptor (suPAR), in a free-ranging population of rhesus macaques, Macaca mulatta. We collected 854 urine samples from 172 adult monkeys and quantified how urinary suPAR and neopterin concentrations were related to affiliative behaviour and agonistic behaviour received over 60 days. In females, but not in males, higher rates of affiliative interactions were associated with lower neopterin concentrations, while conversely, experiencing more agonistic interactions was associated with higher neopterin concentrations. The association between affiliation and neopterin concentration was modulated by age, with older females experiencing a stronger negative association between affiliative behaviour and neopterin concentration. There were no associations between suPAR concentration and social environment for either sex. This study demonstrates that proinflammatory immune activity is a potential mechanism mediating the association between social environment and fitness under naturalistic conditions and that age can be an important modulator of the effect of social environment on the immune system.
Collapse
Affiliation(s)
- Eve B. Cooper
- Department of Anthropology, New York University, New York, NY, U.S.A
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, NY, U.S.A
| | - Connor Whalen
- Department of Anthropology, New York University, New York, NY, U.S.A
| | - Nina Beeby
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, NY, U.S.A
- The Graduate Center of City University of New York, New York, NY, U.S.A
| | | | - Daniel Phillips
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, U.S.A
| | | | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, U.S.A
- School of Life Sciences, Arizona State University, Tempe, AZ, U.S.A
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, U.S.A
- School for Human Evolution and Social Change, Arizona State University, Tempe, AZ, U.S.A
| | - Lauren J. N. Brent
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, U.K
| | - James P. Higham
- Department of Anthropology, New York University, New York, NY, U.S.A
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, NY, U.S.A
| |
Collapse
|
10
|
Francis G, Wang Q. History of Health at Cayo Santiago-An Investigation of Environmental and Genetic Influences on the Skeletal Remains of the Introduced Rhesus Macaque (Macaca mulatta) Colony. Am J Primatol 2025; 87:e23722. [PMID: 39777726 DOI: 10.1002/ajp.23722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 11/22/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
The Cayo Santiago rhesus macaque colony is a renowned primate population that has experienced significant natural and anthropogenic ecological variation in their 85-year history. Demographic and familial information is also tracked and collated for the majority of monkeys. Thus, the health history of rhesus macaques at Cayo Santiago should reflect the impacts of both environmental and genetic factors. In this study, we utilized a sample of skeletal remains comprised of 2787 individuals (1571 females, 1091 males), born between 1938 and 2017 from the derived skeletal collection of the primate colony to assess variation in survivorship, pathology, bone mineral density (BMD), and dental eruption status, in the context of hurricane impacts, nutritional fluctuations, and matriline genealogy. Results demonstrated that rhesus macaques at Cayo Santiago exhibit a range of skeletal pathologies that encompass biomedical and archaeological significance, multiple etiologies, severities, locations, and types, in addition to a secular trend of declining BMD that is hypothesized to reflect decreasing physical activity levels under increasing population densities. Specifically, hurricane impacts were found to increase the rate of systemic disease, decrease BMD in young adults, and delay eruption of the primary dentition. Certain matrilines exhibited heightened levels of systemic disease at early ages while others exhibited greater rates of congenital disease. Early-life adversity, through the experience of major hurricanes, may enhance inflammatory pathways, heightening the risk of disease and accelerating the aging process leading to reduced BMD. Such impacts may underly greater levels of observed infection post-hurricane through intensification of pathogen transmission and disease rates brought on by hurricane-adaptive social strategies that favor closer proximity. Familial susceptibility to disease indicates heritable host genetic factors are likely influencing disease patterning in the population. A cluster of congenital diseases may most convincingly illustrate this, or alternatively reflects low levels of genetic diversity in the population.
Collapse
Affiliation(s)
- George Francis
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Qian Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| |
Collapse
|
11
|
Siracusa ER, Pavez-Fox MA, Negron-Del Valle JE, Phillips D, Platt ML, Snyder-Mackler N, Higham JP, Brent LJN, Silk MJ. Social ageing can protect against infectious disease in a group-living primate. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220462. [PMID: 39463240 PMCID: PMC11528358 DOI: 10.1098/rstb.2022.0462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 10/29/2024] Open
Abstract
The benefits of social living are well established, but sociality also comes with costs, including infectious disease risk. This cost-benefit ratio of sociality is expected to change across individuals' lifespans, which may drive changes in social behaviour with age. To explore this idea, we combine data from a group-living primate for which social ageing has been described with epidemiological models to show that having lower social connectedness when older can protect against the costs of a hypothetical, directly transmitted endemic pathogen. Assuming no age differences in epidemiological characteristics (susceptibility to, severity and duration of infection), older individuals suffered lower infection costs, which was explained largely because they were less connected in their social networks than younger individuals. This benefit of 'social ageing' depended on epidemiological characteristics and was greatest when infection severity increased with age. When infection duration increased with age, social ageing was beneficial only when pathogen transmissibility was low. Older individuals benefited most from having a lower frequency of interactions (strength) and network embeddedness (closeness) and benefited less from having fewer social partners (degree). Our study provides a first examination of the epidemiology of social ageing, demonstrating the potential for pathogens to influence the evolutionary dynamics of social ageing in natural populations.This article is part of the discussion meeting issue 'Understanding age and society using natural populations'.
Collapse
Affiliation(s)
- Erin R. Siracusa
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| | - Melissa A. Pavez-Fox
- Department of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | | | - Daniel Phillips
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Marketing, University of Pennsylvania, Philadelphia, PA, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- School for Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
| | - James P. Higham
- Department of Anthropology, New York University, New York, NY, USA
| | - Lauren J. N. Brent
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| | - Matthew J. Silk
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Melin AD. Baboons, bacteria, and biological clocks address an age-old question. eLife 2024; 13:e104715. [PMID: 39625754 PMCID: PMC11614382 DOI: 10.7554/elife.104715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
Studying the fecal microbiota of wild baboons helps provide new insight into the factors that influence biological aging.
Collapse
Affiliation(s)
- Amanda D Melin
- Department of Anthropology and Archaeology, University of CalgaryCalgaryCanada
| |
Collapse
|
13
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. GeroScience 2024; 46:5819-5841. [PMID: 38509416 PMCID: PMC11493911 DOI: 10.1007/s11357-024-01133-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent subcutaneous bioidentical E2 chronic treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p = 1.6 × 10-51) and upregulation (p = 3.8 × 10-3) of UBE2M across both brain regions provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p = 1.9 × 10-4; interaction p = 3.5 × 10-2) of LTBR in the PFC provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step toward understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Larry Wilhelm
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dongqin Zhu
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Jessica Bodie
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
14
|
Costa CE, Watowich MM, Goldman EA, Sterner KN, Negron-Del Valle JE, Phillips D, Platt ML, Montague MJ, Brent LJN, Higham JP, Snyder-Mackler N, Lea AJ. Genetic Architecture of Immune Cell DNA Methylation in the Rhesus Macaque. Mol Ecol 2024:e17576. [PMID: 39582237 DOI: 10.1111/mec.17576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/23/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Genetic variation that impacts gene regulation, rather than protein function, can have strong effects on trait variation both within and between species. Epigenetic mechanisms, such as DNA methylation, are often an important intermediate link between genotype and phenotype, yet genetic effects on DNA methylation remain understudied in natural populations. To address this gap, we used reduced representation bisulfite sequencing to measure DNA methylation levels at 555,856 CpGs in peripheral whole blood of 573 samples collected from free-ranging rhesus macaques (Macaca mulatta) living on the island of Cayo Santiago, Puerto Rico. We used allele-specific methods to map cis-methylation quantitative trait loci (meQTL) and tested for effects of 243,389 single nucleotide polymorphisms (SNPs) on local DNA methylation levels. Of 776,092 tested SNP-CpG pairs, we identified 516,213 meQTL, with 69.12% of CpGs having at least one meQTL (FDR < 5%). On average, meQTL explained 21.2% of nearby methylation variance, significantly more than age or sex. meQTL were enriched in genomic compartments where methylation is likely to impact gene expression, for example, promoters, enhancers and binding sites for methylation-sensitive transcription factors. In support, using mRNA-seq data from 172 samples, we confirmed 332 meQTL as whole blood cis-expression QTL (eQTL) in the population, and found meQTL-eQTL genes were enriched for immune response functions, like antigen presentation and inflammation. Overall, our study takes an important step towards understanding the genetic architecture of DNA methylation in natural populations, and more generally points to the biological mechanisms driving phenotypic variation in our close relatives.
Collapse
Affiliation(s)
- Christina E Costa
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Marina M Watowich
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Kirstin N Sterner
- Department of Anthropology, University of Oregon, Eugene, Oregon, USA
| | - Josue E Negron-Del Valle
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| | - Daniel Phillips
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - James P Higham
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Noah Snyder-Mackler
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
- Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Amanda J Lea
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Huber HF, Ainsworth HC, Quillen EE, Salmon A, Ross C, Azhar AD, Bales K, Basso MA, Coleman K, Colman R, Darusman HS, Hopkins W, Hotchkiss CE, Jorgensen MJ, Kavanagh K, Li C, Mattison JA, Nathanielsz PW, Saputro S, Scorpio DG, Sosa PM, Vallender EJ, Wang Y, Zeiss CJ, Shively CA, Cox LA. Comparative lifespan and healthspan of nonhuman primate species common to biomedical research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606010. [PMID: 39131309 PMCID: PMC11312576 DOI: 10.1101/2024.07.31.606010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
There is a critical need to generate age- and sex-specific survival curves to characterize chronological aging consistently across nonhuman primates (NHP) used in biomedical research. Sex-specific Kaplan-Meier survival curves were computed in 12 translational aging models: baboon, bonnet macaque, chimpanzee, common marmoset, coppery titi monkey, cotton-top tamarin, cynomolgus macaque, Japanese macaque, pigtail macaque, rhesus macaque, squirrel monkey, and vervet/African green. After employing strict inclusion criteria, primary results are based on 12,269 NHP that survived to adulthood and died of natural/health-related causes. A secondary analysis was completed for 32,616 NHP that died of any cause. Results show a pattern of reduced male survival among catarrhines (African and Asian primates), especially macaques, but not platyrrhines (Central and South American primates). For many species, median lifespans were lower than previously reported. An important consideration is that these analyses may offer a better reflection of healthspan than lifespan since research NHP are typically euthanized for humane welfare reasons before their natural end of life. This resource represents the most comprehensive characterization of sex-specific lifespan and age-at-death distributions for 12 biomedically relevant species, to date. These results clarify relationships among NHP ages and provide a valuable resource for the aging research community, improving human-NHP age equivalencies, informing investigators of expected survival rates, providing a metric for comparisons in future studies, and contributing to understanding of factors driving lifespan differences within and among species.
Collapse
Affiliation(s)
| | | | - Ellen E Quillen
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Adam Salmon
- University of Texas Health Science Center, San Antonio, TX, USA
| | - Corinna Ross
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Adinda D Azhar
- Primate Research Center IPB University, Bogor, Indonesia
| | - Karen Bales
- California National Primate Research Center, Davis, CA, USA
- University of California, Davis, CA, USA
| | - Michele A Basso
- Washington National Primate Research Center, Seattle, WA, USA
| | - Kristine Coleman
- Oregon National Primate Research Center, Hillsboro, OR, USA
- Oregon Health & Science University, Portland, OR, USA
| | - Ricki Colman
- Wisconsin National Primate Research Center, Madison, WI, USA
| | - Huda S Darusman
- Primate Research Center IPB University, Bogor, Indonesia
- School of Veterinary Medicine and Biomedical Sciences IPB University, Bogor, Indonesia
| | - William Hopkins
- The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | | | | | - Kylie Kavanagh
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
- University of Tasmania, Hobart, Tasmania, Australia
| | - Cun Li
- University of Wyoming, Laramie, WY, USA
| | - Julie A Mattison
- National Institute on Aging, National Institutes of Health, Gaithersburg, MD, USA
| | - Peter W Nathanielsz
- Texas Biomedical Research Institute, San Antonio, TX, USA
- University of Wyoming, Laramie, WY, USA
| | - Suryo Saputro
- Primate Research Center IPB University, Bogor, Indonesia
| | - Diana G Scorpio
- Texas Biomedical Research Institute, San Antonio, TX, USA
- Envol Biomedical, Immokalee, FL, USA
| | | | - Eric J Vallender
- Tulane National Primate Research Center, Covington, LA, USA
- New England Primate Research Center, Southborough, MA, USA
| | - Yaomin Wang
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Carol A Shively
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Laura A Cox
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
16
|
Zhou J, Chen Y, Jin X, Mao W, Xiao Z, Zhang S, Zhang T, Liu T, Kendrick K, Jiang X. Fusing multi-scale functional connectivity patterns via Multi-Branch Vision Transformer (MB-ViT) for macaque brain age prediction. Neural Netw 2024; 179:106592. [PMID: 39168070 DOI: 10.1016/j.neunet.2024.106592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 06/03/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024]
Abstract
Brain age (BA) is defined as a measure of brain maturity and could help characterize both the typical brain development and neuropsychiatric disorders in mammals. Various biological phenotypes have been successfully applied to predict BA of human using chronological age (CA) as label. However, whether the BA of macaque, one of the most important animal models, can also be reliably predicted is largely unknown. To address this question, we propose a novel deep learning model called Multi-Branch Vision Transformer (MB-ViT) to fuse multi-scale (i.e., from coarse-grained to fine-grained) brain functional connectivity (FC) patterns derived from resting state functional magnetic resonance imaging (rs-fMRI) data to predict BA of macaques. The discriminative functional connections and the related brain regions contributing to the prediction are further identified based on Gradient-weighted Class Activation Mapping (Grad-CAM) method. Our proposed model successfully predicts BA of 450 normal rhesus macaques from the publicly available PRIMatE Data Exchange (PRIME-DE) dataset with lower mean absolute error (MAE) and mean square error (MSE) as well as higher Pearson's correlation coefficient (PCC) and coefficient of determination (R2) compared to other baseline models. The correlation between the predicted BA and CA reaches as high as 0.82 of our proposed method. Furthermore, our analysis reveals that the functional connections predominantly contributing to the prediction results are situated in the primary motor cortex (M1), visual cortex, area v23 in the posterior cingulate cortex, and dysgranular temporal pole. In summary, our proposed deep learning model provides an effective tool to accurately predict BA of primates (macaque in this study), and lays a solid foundation for future studies of age-related brain diseases in those animal models.
Collapse
Affiliation(s)
- Jingchao Zhou
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuzhong Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuewei Jin
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Mao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenxiang Xiao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Songyao Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Tuo Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Tianming Liu
- School of Computing, University of Georgia, Athens, USA
| | - Keith Kendrick
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xi Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
17
|
Watowich MM, Costa CE, Chiou KL, Goldman EA, Petersen RM, Patterson S, Martínez MI, Sterner KN, Horvath JE, Montague MJ, Platt ML, Brent LJN, Higham JP, Lea AJ, Snyder-Mackler N. Immune gene regulation is associated with age and environmental adversity in a nonhuman primate. Mol Ecol 2024; 33:e17445. [PMID: 39032090 PMCID: PMC11521774 DOI: 10.1111/mec.17445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/27/2024] [Accepted: 06/14/2024] [Indexed: 07/22/2024]
Abstract
Phenotypic aging is ubiquitous across mammalian species, suggesting shared underlying mechanisms of aging. Aging is linked to molecular changes to DNA methylation and gene expression, and environmental factors, such as severe external challenges or adversities, can moderate these age-related changes. Yet, it remains unclear whether environmental adversities affect gene regulation via the same molecular pathways as chronological, or 'primary', aging. Investigating molecular aging in naturalistic animal populations can fill this gap by providing insight into shared molecular mechanisms of aging and the effects of a greater diversity of environmental adversities - particularly those that can be challenging to study in humans or laboratory organisms. Here, we characterised molecular aging - specifically, CpG methylation - in a sample of free-ranging rhesus macaques living off the coast of Puerto Rico (n samples = 571, n individuals = 499), which endured a major hurricane during our study. Age was associated with methylation at 78,661 sites (31% of all sites tested). Age-associated hypermethylation occurred more frequently in areas of active gene regulation, while hypomethylation was enriched in regions that show less activity in immune cells, suggesting these regions may become de-repressed in older individuals. Age-associated hypomethylation also co-occurred with increased chromatin accessibility while hypermethylation showed the opposite trend, hinting at a coordinated, multi-level loss of epigenetic stability during aging. We detected 32,048 CpG sites significantly associated with exposure to a hurricane, and these sites overlapped age-associated sites, most strongly in regulatory regions and most weakly in quiescent regions. Together, our results suggest that environmental adversity may contribute to aging-related molecular phenotypes in regions of active gene transcription, but that primary aging has specific signatures in non-regulatory regions.
Collapse
Affiliation(s)
- Marina M. Watowich
- Department of Biology, University of Washington, Seattle, Washington, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | - Christina E. Costa
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Kenneth L. Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Elisabeth A. Goldman
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Rachel M. Petersen
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | - Sam Patterson
- Department of Anthropology, New York University, New York, New York, USA
| | | | - Melween I. Martínez
- Caribbean Primate Research Center, Unit of Comparative Medicine, University of Puerto Rico, San Juan, Puerto Rico, USA
| | | | - Julie E. Horvath
- Research and Collections Section, North Carolina Museum of Natural Sciences, Raleigh, North Carolina, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Renaissance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael J. Montague
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael L. Platt
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Marketing Department, Wharton School of Business, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lauren J. N. Brent
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| | - James P. Higham
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Amanda J. Lea
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
- Child and Brain Development, Canadian Institute for Advanced Research, Toronto, Canada
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
- Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
18
|
Kohama SG, Urbanski HF. The aged female rhesus macaque as a translational model for human menopause and hormone therapy. Horm Behav 2024; 166:105658. [PMID: 39531811 PMCID: PMC11602343 DOI: 10.1016/j.yhbeh.2024.105658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Progress in understanding the causes of physiological and behavioral changes in post-menopausal women is hampered by the paucity of animal models that accurately recapitulate these age-associated changes. Here we evaluate the translational potential of female rhesus macaques (Macaca mulatta). Like women, these long-lived diurnal primates show marked neuroendocrine changes during aging, as well as perturbed sleep-wake cycles and cognitive decline. Furthermore, the brains of old rhesus macaques show some of the same pathological hallmarks of Alzheimer's disease as do humans, including amyloidosis and tauopathology. Importantly, unlike humans, rhesus macaques can be maintained under tightly controlled environmental conditions, such as photoperiod, temperature and diet, and tissues can be collected with zero postmortem interval; this makes them especially suitable for studies aimed at elucidating underlying molecular mechanisms. Recent findings from female macaques are helping to elucidate how sex-steroids influence gene expression within the brain and contribute to the maintenance of cognitive function and amelioration of age-associated pathologies. Taken together, these findings emphasize the translational value of female rhesus macaques as a model for elucidating causal mechanisms that underlie normative and pathological changes in post-menopausal women. They also provide a pragmatic platform upon which to develop safe and effective therapies.
Collapse
Affiliation(s)
- Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| |
Collapse
|
19
|
Siracusa ER, Pavez-Fox MA, Negron-Del Valle JE, Phillips D, Platt ML, Snyder-Mackler N, Higham JP, Brent LJN, Silk MJ. Social ageing can protect against infectious disease in a group-living primate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.09.584237. [PMID: 38559098 PMCID: PMC10979879 DOI: 10.1101/2024.03.09.584237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The benefits of social living are well established, but sociality also comes with costs, including infectious disease risk. This cost-benefit ratio of sociality is expected to change across individuals' lifespans, which may drive changes in social behaviour with age. To explore this idea, we combine data from a group-living primate for which social ageing has been described with epidemiological models to show that having lower social connectedness when older can protect against the costs of a hypothetical, directly transmitted endemic pathogen. Assuming no age differences in epidemiological characteristics (susceptibility to, severity, and duration of infection), older individuals suffered lower infection costs, which was explained largely because they were less connected in their social networks than younger individuals. This benefit of 'social ageing' depended on epidemiological characteristics and was greatest when infection severity increased with age. When infection duration increased with age, social ageing was beneficial only when pathogen transmissibility was low. Older individuals benefited most from having a lower frequency of interactions (strength) and network embeddedness (closeness) and benefited less from having fewer social partners (degree). Our study provides a first examination of the epidemiology of social ageing, demonstrating the potential for pathogens to influence evolutionary dynamics of social ageing in natural populations.
Collapse
Affiliation(s)
- Erin R. Siracusa
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| | | | | | - Daniel Phillips
- Center for Evolution and Medicine, Arizona State University, Arizona, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, PA, USA
- Department of Psychology, University of Pennsylvania, PA, USA
- Department of Marketing, University of Pennsylvania, PA, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Arizona, USA
- School of Life Sciences, Arizona State University, Arizona, USA
- School for Human Evolution and Social Change, Arizona State University, Arizona, USA
| | - James P. Higham
- Department of Anthropology, New York University, New York, USA
| | - Lauren J. N. Brent
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| | - Matthew J. Silk
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Kelberman MA, Rodberg E, Arabzadeh E, Bair-Marshall CJ, Berridge CW, Berrocoso E, Breton-Provencher V, Chandler DJ, Che A, Davy O, Devilbiss DM, Downs AM, Drummond G, Dvorkin R, Fazlali Z, Froemke RC, Glennon E, Gold JI, Ito H, Jiang X, Johansen JP, Kaye AP, Kim JR, Kuo CC, Liu RJ, Liu Y, Llorca-Torralba M, McCall JG, McElligott ZA, McKinney AM, Miguelez C, Min MY, Nowlan AC, Omrani M, Poe GR, Pickering AE, Ranjbar-Slamloo Y, Razquin J, Rodenkirch C, Sales AC, Satyasambit R, Shea SD, Sur M, Tkaczynski JA, Torres-Sanchez S, Uematsu A, Vazquez CR, Vreven A, Wang Q, Waterhouse BD, Yang HW, Yang JH, Zhao L, Zouridis IS, Weinshenker D, Vazey E, Totah NK. Diversity of ancestral brainstem noradrenergic neurons across species and multiple biological factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618224. [PMID: 39464004 PMCID: PMC11507722 DOI: 10.1101/2024.10.14.618224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The brainstem region, locus coeruleus (LC), has been remarkably conserved across vertebrates. Evolution has woven the LC into wide-ranging neural circuits that influence functions as broad as autonomic systems, the stress response, nociception, sleep, and high-level cognition among others. Given this conservation, there is a strong possibility that LC activity is inherently similar across species, and furthermore that age, sex, and brain state influence LC activity similarly across species. The degree to which LC activity is homogenous across these factors, however, has never been assessed due to the small sample size of individual studies. Here, we pool data from 20 laboratories (1,855 neurons) and show diversity across both intrinsic and extrinsic factors such as species, age, sex and brain state. We use a negative binomial regression model to compare activity from male monkeys, and rats and mice of both sexes that were recorded across brain states from brain slices ex vivo or under different anesthetics or during wakefulness in vivo. LC activity differed due to complex interactions of species, sex, and brain state. The LC became more active during aging, independent of sex. Finally, in contrast to the foundational principle that all species express two distinct LC firing modes ("tonic" or "phasic"), we discovered great diversity within spontaneous LC firing patterns. Different factors were associated with higher incidence of some firing modes. We conclude that the activity of the evolutionarily-ancient LC is not conserved. Inherent differences due to age and species-sex-brain state interactions have implications for understanding the role of LC in species-specific naturalistic behavior, as well as in psychiatric disorders, cardiovascular disease, immunology, and metabolic disorders.
Collapse
Affiliation(s)
- Michael A. Kelberman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Ellen Rodberg
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Ehsan Arabzadeh
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, AUS
| | - Chloe J. Bair-Marshall
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Craig W. Berridge
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Esther Berrocoso
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Alicia Che
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Oscar Davy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | | | - Anthony M. Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle Drummond
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roman Dvorkin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Zeinab Fazlali
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- Department of Psychiatry, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Robert C. Froemke
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Otolaryngology, NYU Grossman School of Medicine, New York, NY, USA
| | - Erin Glennon
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Neurology, Weill Cornell Medicine, New York
| | - Joshua I. Gold
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Hiroki Ito
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Xiaolong Jiang
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
- Department of Ophthalmology, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | | | - Alfred P. Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, USA
| | - Jenny R. Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Rong-Jian Liu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yang Liu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordan G. McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew M. McKinney
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Alexandra C. Nowlan
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohsen Omrani
- Department of Psychiatry, Queen’s University, Kingston, ON, Canada
| | - Gina R. Poe
- Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Anthony Edward Pickering
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Yadollah Ranjbar-Slamloo
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Jone Razquin
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Charles Rodenkirch
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Anna C. Sales
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Rath Satyasambit
- RIKEN Center for Brain Science, Wako-shi Saitama, Japan
- Department of Computer Science, Tokyo Institute of Technology, Midori, Yokohama, Japan
| | | | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Sonia Torres-Sanchez
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Akira Uematsu
- Human Informatics and Information Research Institute, National Institute of Advanced Industrial Science and Technology, Japan
| | - Chayla R. Vazquez
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Amelien Vreven
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Hsiu-Wen Yang
- Department of Biomedical Sciences, Chung-Shan Medical University, Taichung, Taiwan
| | - Jen-Hau Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Ioannis S. Zouridis
- Graduate Training Centre of Neuroscience, International Max Planck Research School (IMPRS), University of Tübingen, Tübingen, Germany
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | | | - Elena Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Nelson K. Totah
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| |
Collapse
|
21
|
Lepore G, Succu S, Cappai MG, Frau A, Senes A, Zedda M, Farina V, Gadau SD. Morphological and Metabolic Features of Brain Aging in Rodents, Ruminants, Carnivores, and Non-Human Primates. Animals (Basel) 2024; 14:2900. [PMID: 39409849 PMCID: PMC11482532 DOI: 10.3390/ani14192900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Brain aging in mammals is characterized by morphological and functional changes in neural cells. Macroscopically, this process, leading to progressive cerebral volume loss and functional decline, includes memory and motor neuron deficits, as well as behavioral disorders. Morphologically, brain aging is associated with aged neurons and astrocytes, appearing enlarged and flattened, and expressing enhanced pH-dependent β-galactosidase activity. Multiple mechanisms are considered hallmarks of cellular senescence in vitro, including cell cycle arrest, increased lysosomal activity, telomere shortening, oxidative stress, and DNA damage. The most common markers for senescence identification were identified in (i) proteins implicated in cell cycle arrest, such as p16, p21, and p53, (ii) increased lysosomal mass, and (iii) increased reactive oxygen species (ROS) and senescence-associated secretory phenotype (SASP) expression. Finally, dysfunctional autophagy, a process occurring during aging, contributes to altering brain homeostasis. The brains of mammals can be studied at cellular and subcellular levels to elucidate the mechanisms on the basis of age-related and degenerative disorders. The aim of this review is to summarize and update the most recent knowledge about brain aging through a comparative approach, where similarities and differences in some mammalian species are considered.
Collapse
Affiliation(s)
- Gianluca Lepore
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy; (S.S.); (M.G.C.); (A.F.); (A.S.); (M.Z.); (V.F.); (S.D.G.)
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Covarrubias BV, Kamminga JM, Muchlinski MN, Munds RA, Villero NV, Bauman SS, Martinez MI, Montague MJ, Higham JP, Melin AD, Veilleux CC. Investigating mechanoreceptor variability and morphometric proxies in Rhesus Macaques: Implications for primate precision touch studies. Anat Rec (Hoboken) 2024:10.1002/ar.25587. [PMID: 39367664 PMCID: PMC11968448 DOI: 10.1002/ar.25587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/26/2024] [Accepted: 09/13/2024] [Indexed: 10/06/2024]
Abstract
The origin of primates has long been associated with an increased emphasis on manual grasping and touch. Precision touch, facilitated by specialized mechanoreceptors in glabrous skin, provides critical sensory feedback for grasping-related tasks and perception of ecologically-relevant stimuli. Despite its importance, studies of mechanoreceptors in primate hands are limited, in part due to challenges of sample availability and histological methods. Dermatoglyphs have been proposed as alternative proxies of mechanoreceptor density. We investigated the relationships between mechanoreceptors (Meissner and Pacinian corpuscles), dermatoglyphs, and demography in the apical finger pads of 15 juvenile to adult rhesus macaques (Macaca mulatta) from a free-ranging population at Cayo Santiago Primate Field Station (Puerto Rico). Our results indicate substantial interindividual variation in mechanoreceptor density (Meissner corpuscles: 11.9-43.3 corpuscles/mm2; Pacinian corpuscles: 0-4.5 corpuscles/mm2). While sex and digit were generally not associated with variation, there was strong evidence of a developmental effect. Specifically, apical pad length, Meissner corpuscle size, and Pacinian corpuscle depth increased while mechanoreceptor densities decreased throughout juvenescence, suggesting that primate mechanoreceptors change as fingers grow during adolescence and then stabilize at physical maturity. We also found Meissner corpuscle density was significantly associated with dermatoglyph ridge width and spacing, such that density predicted by a dermatoglyph model was strongly correlated with observed values. Dermatoglyphs thus offer a useful proxy of relative Meissner corpuscle density in primates, which opens exciting avenues of noninvasive research. Finally, our results underscore the importance of considering demographic factors and methodology in comparative studies of primate touch.
Collapse
Affiliation(s)
| | - Jordan M. Kamminga
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ
| | - MN Muchlinski
- Anatomical Science Education Center, Oregon Health and Science University, Portland, OR
| | - RA Munds
- Department of Anthropology & Archaeology, University of Calgary, Calgary, AB
| | - Núñez V Villero
- Caribbean Primate Research Center, University of Puerto Rico
| | | | | | - MI Martinez
- Caribbean Primate Research Center, University of Puerto Rico
| | - MJ Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA
| | - JP Higham
- Department of Anthropology, New York University, New York, NY
| | - AD Melin
- Department of Anthropology & Archaeology, University of Calgary, Calgary, AB
- Department of Medical Genetics, University of Calgary, Calgary, AB
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB
| | - CC Veilleux
- Department of Anatomy, Midwestern University, Glendale, AZ
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ
| |
Collapse
|
23
|
Longtin A, Watowich MM, Sadoughi B, Petersen RM, Brosnan SF, Buetow K, Cai Q, Gurven MD, Highland HM, Huang YT, Kaplan H, Kraft TS, Lim YAL, Long J, Melin AD, Roberson J, Ng KS, Stieglitz J, Trumble BC, Venkataraman VV, Wallace IJ, Wu J, Snyder-Mackler N, Jones A, Bick AG, Lea AJ. Cost-effective solutions for high-throughput enzymatic DNA methylation sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612068. [PMID: 39314398 PMCID: PMC11419010 DOI: 10.1101/2024.09.09.612068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Characterizing DNA methylation patterns is important for addressing key questions in evolutionary biology, geroscience, and medical genomics. While costs are decreasing, whole-genome DNA methylation profiling remains prohibitively expensive for most population-scale studies, creating a need for cost-effective, reduced representation approaches (i.e., assays that rely on microarrays, enzyme digests, or sequence capture to target a subset of the genome). Most common whole genome and reduced representation techniques rely on bisulfite conversion, which can damage DNA resulting in DNA loss and sequencing biases. Enzymatic methyl sequencing (EM-seq) was recently proposed to overcome these issues, but thorough benchmarking of EM-seq combined with cost-effective, reduced representation strategies has not yet been performed. To do so, we optimized Targeted Methylation Sequencing protocol (TMS)-which profiles ∼4 million CpG sites-for miniaturization, flexibility, and multispecies use at a cost of ∼$80. First, we tested modifications to increase throughput and reduce cost, including increasing multiplexing, decreasing DNA input, and using enzymatic rather than mechanical fragmentation to prepare DNA. Second, we compared our optimized TMS protocol to commonly used techniques, specifically the Infinium MethylationEPIC BeadChip (n=55 paired samples) and whole genome bisulfite sequencing (n=6 paired samples). In both cases, we found strong agreement between technologies (R² = 0.97 and 0.99, respectively). Third, we tested the optimized TMS protocol in three non-human primate species (rhesus macaques, geladas, and capuchins). We captured a high percentage (mean=77.1%) of targeted CpG sites and produced methylation level estimates that agreed with those generated from reduced representation bisulfite sequencing (R² = 0.98). Finally, we applied our protocol to profile age-associated DNA methylation variation in two subsistence-level populations-the Tsimane of lowland Bolivia and the Orang Asli of Peninsular Malaysia-and found age-methylation patterns that were strikingly similar to those reported in high income cohorts, despite known differences in age-health relationships between lifestyle contexts. Altogether, our optimized TMS protocol will enable cost-effective, population-scale studies of genome-wide DNA methylation levels across human and non-human primate species.
Collapse
|
24
|
Bartolomucci A, Kane AE, Gaydosh L, Razzoli M, McCoy BM, Ehninger D, Chen BH, Howlett SE, Snyder-Mackler N. Animal Models Relevant for Geroscience: Current Trends and Future Perspectives in Biomarkers, and Measures of Biological Aging. J Gerontol A Biol Sci Med Sci 2024; 79:glae135. [PMID: 39126297 PMCID: PMC11316208 DOI: 10.1093/gerona/glae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Indexed: 08/12/2024] Open
Abstract
For centuries, aging was considered inevitable and immutable. Geroscience provides the conceptual framework to shift this focus toward a new view that regards aging as an active biological process, and the biological age of an individual as a modifiable entity. Significant steps forward have been made toward the identification of biomarkers for and measures of biological age, yet knowledge gaps in geroscience are still numerous. Animal models of aging are the focus of this perspective, which discusses how experimental design can be optimized to inform and refine the development of translationally relevant measures and biomarkers of biological age. We provide recommendations to the field, including: the design of longitudinal studies in which subjects are deeply phenotyped via repeated multilevel behavioral/social/molecular assays; the need to consider sociobehavioral variables relevant for the species studied; and finally, the importance of assessing age of onset, severity of pathologies, and age-at-death. We highlight approaches to integrate biomarkers and measures of functional impairment using machine learning approaches designed to estimate biological age as well as to predict future health declines and mortality. We expect that advances in animal models of aging will be crucial for the future of translational geroscience but also for the next chapter of medicine.
Collapse
Affiliation(s)
- Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Alice E Kane
- Institute for Systems Biology, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Lauren Gaydosh
- Department of Sociology, University of Texas at Austin, Austin, Texas, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brianah M McCoy
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Brian H Chen
- California Pacific Medical Center Research Institute, Sutter Health, San Francisco, CA, 94143, USA
| | - Susan E Howlett
- Departments of Pharmacology and Medicine (Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Noah Snyder-Mackler
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
25
|
Alldritt S, Ramirez J, de Wael RV, Bethlehem R, Seidlitz J, Wang Z, Nenning K, Esper N, Smallwood J, Franco A, Byeon K, Alexander-Bloch A, Amaral D, Amiez C, Balezeau F, Baxter M, Becker G, Bennett J, Berkner O, Blezer E, Brambrink A, Brochier T, Butler B, Campos L, Canet-Soulas E, Chalet L, Chen A, Cléry J, Constantinidis C, Cook D, Dehaene S, Dorfschmidt L, Drzewiecki C, Erdman J, Everling S, Falchier A, Fleysher L, Fox A, Freiwald W, Froesel M, Froudist-Walsh S, Fudge J, Funck T, Gacoin M, Gale D, Gallivan J, Garin C, Griffiths T, Guedj C, Hadj-Bouziane F, Hamed S, Harel N, Hartig R, Hiba B, Howell B, Jarraya B, Jung B, Kalin N, Karpf J, Kastner S, Klink C, Kovacs-Balint Z, Kroenke C, Kuchan M, Kwok S, Lala K, Leopold D, Li G, Lindenfors P, Linn G, Mars R, Masiello K, Menon R, Messinger A, Meunier M, Mok K, Morrison J, Nacef J, Nagy J, Neudecker V, Neuringer M, Noonan M, Ortiz-Rios M, Perez-Zoghbi J, Petkov C, Pinsk M, Poirier C, Procyk E, Rajimehr R, Reader S, Rudko D, Rushworth M, Russ B, Sallet J, Sanchez M, Schmid M, Schwiedrzik C, Scott J, Sein J, Sharma K, et alAlldritt S, Ramirez J, de Wael RV, Bethlehem R, Seidlitz J, Wang Z, Nenning K, Esper N, Smallwood J, Franco A, Byeon K, Alexander-Bloch A, Amaral D, Amiez C, Balezeau F, Baxter M, Becker G, Bennett J, Berkner O, Blezer E, Brambrink A, Brochier T, Butler B, Campos L, Canet-Soulas E, Chalet L, Chen A, Cléry J, Constantinidis C, Cook D, Dehaene S, Dorfschmidt L, Drzewiecki C, Erdman J, Everling S, Falchier A, Fleysher L, Fox A, Freiwald W, Froesel M, Froudist-Walsh S, Fudge J, Funck T, Gacoin M, Gale D, Gallivan J, Garin C, Griffiths T, Guedj C, Hadj-Bouziane F, Hamed S, Harel N, Hartig R, Hiba B, Howell B, Jarraya B, Jung B, Kalin N, Karpf J, Kastner S, Klink C, Kovacs-Balint Z, Kroenke C, Kuchan M, Kwok S, Lala K, Leopold D, Li G, Lindenfors P, Linn G, Mars R, Masiello K, Menon R, Messinger A, Meunier M, Mok K, Morrison J, Nacef J, Nagy J, Neudecker V, Neuringer M, Noonan M, Ortiz-Rios M, Perez-Zoghbi J, Petkov C, Pinsk M, Poirier C, Procyk E, Rajimehr R, Reader S, Rudko D, Rushworth M, Russ B, Sallet J, Sanchez M, Schmid M, Schwiedrzik C, Scott J, Sein J, Sharma K, Shmuel A, Styner M, Sullivan E, Thiele A, Todorov O, Tsao D, Tusche A, Vlasova R, Wang Z, Wang L, Wang J, Weiss A, Wilson C, Yacoub E, Zarco W, Zhou Y, Zhu J, Margulies D, Fair D, Schroeder C, Milham M, Xu T. Brain Charts for the Rhesus Macaque Lifespan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610193. [PMID: 39257737 PMCID: PMC11383706 DOI: 10.1101/2024.08.28.610193] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Recent efforts to chart human brain growth across the lifespan using large-scale MRI data have provided reference standards for human brain development. However, similar models for nonhuman primate (NHP) growth are lacking. The rhesus macaque, a widely used NHP in translational neuroscience due to its similarities in brain anatomy, phylogenetics, cognitive, and social behaviors to humans, serves as an ideal NHP model. This study aimed to create normative growth charts for brain structure across the macaque lifespan, enhancing our understanding of neurodevelopment and aging, and facilitating cross-species translational research. Leveraging data from the PRIMatE Data Exchange (PRIME-DE) and other sources, we aggregated 1,522 MRI scans from 1,024 rhesus macaques. We mapped non-linear developmental trajectories for global and regional brain structural changes in volume, cortical thickness, and surface area over the lifespan. Our findings provided normative charts with centile scores for macaque brain structures and revealed key developmental milestones from prenatal stages to aging, highlighting both species-specific and comparable brain maturation patterns between macaques and humans. The charts offer a valuable resource for future NHP studies, particularly those with small sample sizes. Furthermore, the interactive open resource (https://interspeciesmap.childmind.org) supports cross-species comparisons to advance translational neuroscience research.
Collapse
Affiliation(s)
- S. Alldritt
- Center for the Integrative Developmental Neuroscience, Child Mind Institute
| | | | | | - R. Bethlehem
- University of Cambridge, Department of Psychology
| | | | | | | | | | | | | | | | - A. Alexander-Bloch
- Department of Child and Adolescent Psychiatry and Behavioral Science, Children’s Hospital of Philadelphia
- Department of Psychiatry, University of Pennsylvania
| | - D.G. Amaral
- Department of Psychiatry and Behavioral Sciences and The MIND Institute
- University of California Davis
| | - C. Amiez
- Stem Cell and Brain Research Institute
| | | | - M.G. Baxter
- Section on Comparative Medicine, Wake Forest University School of Medicine
| | | | - J. Bennett
- University of California Davis, Dept of Psychology
| | - O. Berkner
- Translational Neuroscience division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
| | | | | | | | - B. Butler
- Translational Neuroscience Division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
| | | | | | | | - A. Chen
- East China Normal University
| | | | | | | | | | | | | | | | | | - A. Falchier
- Translational Neuroscience Division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
| | | | - A. Fox
- University of California Davis
| | | | - M. Froesel
- Institute for Cognitive Science Marc Jeannerod
| | | | | | | | - M. Gacoin
- Institute for Cognitive Science Marc Jeannerod
| | | | | | - C.M. Garin
- Department of Biomedical Engineering, Vanderbilt University
- Institut des Sciences Cognitives Marc Jeannerod (ISC-MJ)
| | | | - C. Guedj
- Lyon Neuroscience Research Center, University of Geneva
| | | | - S.B. Hamed
- Institute for Cognitive Science Marc Jeannerod
| | | | - R. Hartig
- Translational Neuroscience division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
| | - B. Hiba
- Institute for Cognitive Science Marc Jeannerod
| | - B.R. Howell
- Emory National Primate Research Center, Emory University
- Fralin Biomedical Research Institute, Virginia Tech
- Carilion Department of Human Development and Family Science, Virginia Tech
| | | | | | | | - J. Karpf
- Oregon National Primate Research Center
| | - S. Kastner
- Princeton Neuroscience Institute & Department of Psychology, Princeton University
| | - C. Klink
- Netherlands Institute for Neuroscience
| | | | | | | | | | - K.N. Lala
- Centre for Social Learning and Cognitive Evolution, School of Biology, University of St. Andrews
| | | | - G. Li
- University of North Carolina at Chapel Hill
| | - P. Lindenfors
- Institute for Futures Studies, Stockholm, Sweden
- Centre for Cultural Evolution & Department of Zoology, Stockholm University, Sweden
| | - G. Linn
- Translational Neuroscience division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
| | | | - K. Masiello
- Translational Neuroscience division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
| | | | | | - M. Meunier
- Lyon Neuroscience Research Center, ImpAct Team
| | | | | | | | - J. Nagy
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai
| | | | | | | | - M. Ortiz-Rios
- Functional Imaging Laboratory, German Primate Center – Leibniz Institute for Primate Research
| | | | | | - M. Pinsk
- Princeton Neuroscience Institute, Princeton University
| | | | - E. Procyk
- Stem Cell and Brain Research Institute
| | - R. Rajimehr
- McGovern Institute for Brain Research, Massachusetts Institute of Technology
| | - S.M. Reader
- Department of Biology, Utrecht University
- Department of Biology, McGill University
| | | | | | - B.E. Russ
- Translational Neuroscience division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
| | - J. Sallet
- University of Oxford
- INSERM Stem Cell & Brain Research Institute
| | - M.M. Sanchez
- Emory National Primate Research Center; Emory University
- Department of Psychiatry & Behavioral Sciences, School of Medicine
| | | | - C.M. Schwiedrzik
- Ruhr University Bochum, Faculty of Biology and Biotechnology, Cognitive Neurobiology
- Neural Circuits and Cognition Lab, European Neuroscience Institute Göttingen
- Perception and Plasticity Group, German Primate Center – Leibniz Institute for Primate Research
| | - J.A. Scott
- Department of Bioengineering, Santa Clara University
| | | | | | | | - M. Styner
- University of North Carolina at Chapel Hill
| | | | | | - O.S. Todorov
- Department of Biology and Helmholtz Institute, Utrecht University
| | - D. Tsao
- Department of Computation and Neural Systems, California Institute of Technology
| | | | - R. Vlasova
- University of North Carolina at Chapel Hill
| | | | - L. Wang
- East China Normal University
| | - J. Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | | | | | | | | | - Y. Zhou
- Krieger Mind/Brain Institute, Department of Neurosurgery, Johns Hopkins University
| | - J. Zhu
- Department of Biomedical Engineering, Vanderbilt University
| | | | | | - C. Schroeder
- Translational Neuroscience division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute
- Deptartment of Psychiatry, Neurology and Neurosurgery, Columbia University
| | - M. Milham
- Child Mind Institute
- Nathan Kline Institute
| | - T. Xu
- Center for the Integrative Developmental Neuroscience, Child Mind Institute
| |
Collapse
|
26
|
Frye BM, Negrey JD, Johnson CSC, Kim J, Barcus RA, Lockhart SN, Whitlow CT, Chiou KL, Snyder-Mackler N, Montine TJ, Craft S, Shively CA, Register TC. Mediterranean diet protects against a neuroinflammatory cortical transcriptome: Associations with brain volumetrics, peripheral inflammation, social isolation, and anxiety in nonhuman primates (Macaca fascicularis). Brain Behav Immun 2024; 119:681-692. [PMID: 38636565 DOI: 10.1016/j.bbi.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/17/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Mediterranean diets may be neuroprotective and prevent cognitive decline relative to Western diets; however, the underlying biology is poorly understood. We assessed the effects of Western versus Mediterranean-like diets on RNAseq-generated transcriptional profiles in lateral temporal cortex and their relationships with longitudinal changes in neuroanatomy, circulating monocyte gene expression, and observations of social isolation and anxiety in 38 socially-housed, middle-aged female cynomolgus macaques (Macaca fascicularis). Diet resulted in differential expression of seven transcripts (FDR < 0.05). Cyclin dependent kinase 14 (CDK14), a proinflammatory regulator, was lower in the Mediterranean group. The remaining six transcripts [i.e., "lunatic fringe" (LFNG), mannose receptor C type 2 (MRC2), solute carrier family 3 member 2 (SLCA32), butyrophilin subfamily 2 member A1 (BTN2A1), katanin regulatory subunit B1 (KATNB1), and transmembrane protein 268 (TMEM268)] were higher in cortex of the Mediterranean group and generally associated with anti-inflammatory/neuroprotective pathways. KATNB1 encodes a subcomponent of katanin, important in maintaining microtubule homeostasis. BTN2A1 is involved in immunomodulation of γδ T-cells which have anti-neuroinflammatory and neuroprotective effects. CDK14, LFNG, MRC2, and SLCA32 are associated with inflammatory pathways. The latter four differentially expressed cortex transcripts were associated with peripheral monocyte transcript levels, neuroanatomical changes determined by MRI, and with social isolation and anxiety. These results provide important insights into the potential mechanistic processes linking diet, peripheral and central inflammation, and behavior. Collectively, our results provide evidence that, relative to Western diets, Mediterranean diets confer protection against peripheral and central inflammation which is reflected in preserved brain structure and socioemotional behavior. Ultimately, such protective effects may confer resilience to the development of neuropathology and associated disease.
Collapse
Affiliation(s)
- Brett M Frye
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Biology, Emory and Henry College, Emory, VA, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA
| | - Jacob D Negrey
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; School of Anthropology, University of Arizona, Tucson, AZ, USA
| | | | - Jeongchul Kim
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Richard A Barcus
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Samuel N Lockhart
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher T Whitlow
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kenneth L Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA; School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
| | | | - Suzanne Craft
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carol A Shively
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA.
| | - Thomas C Register
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA.
| |
Collapse
|
27
|
Cho HM, Choe SH, Lee JR, Park HR, Ko MG, Lee YJ, Lee HY, Park SH, Park SJ, Kim YH, Huh JW. Transcriptome analysis of cynomolgus macaques throughout their lifespan reveals age-related immune patterns. NPJ AGING 2024; 10:30. [PMID: 38902280 PMCID: PMC11189941 DOI: 10.1038/s41514-024-00158-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Despite the different perspectives by diverse research sectors spanning several decades, aging research remains uncharted territory for human beings. Therefore, we investigated the transcriptomic characteristics of eight male healthy cynomolgus macaques, and the annual sampling was designed with two individuals in four age groups. As a laboratory animal, the macaques were meticulously shielded from all environmental factors except aging. The results showed recent findings of certain immune response and the age-associated network of primate immunity. Three important aging patterns were identified and each gene clusters represented a different immune response. The increased expression pattern was predominantly associated with innate immune cells, such as Neutrophils and NK cells, causing chronic inflammation with aging whereas the other two decreased patterns were associated with adaptive immunity, especially "B cell activation" affecting antibody diversity of aging. Furthermore, the hub gene network of the patterns reflected transcriptomic age and correlated with human illness status, aiding in future human disease prediction. Our macaque transcriptome profiling results offer systematic insights into the age-related immunological features of primates.
Collapse
Affiliation(s)
- Hyeon-Mu Cho
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Se-Hee Choe
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
| | - Ja-Rang Lee
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56216, Republic of Korea
| | - Hye-Ri Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Min-Gyeong Ko
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Yun-Jung Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea
| | - Hwal-Yong Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
| | - Sung Hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea
| | - Sang-Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science & Technology (UST), Cheongju, 28116, Republic of Korea.
| |
Collapse
|
28
|
Charbonneau JA, Santistevan AC, Raven EP, Bennett JL, Russ BE, Bliss-Moreau E. Evolutionarily conserved neural responses to affective touch in monkeys transcend consciousness and change with age. Proc Natl Acad Sci U S A 2024; 121:e2322157121. [PMID: 38648473 PMCID: PMC11067024 DOI: 10.1073/pnas.2322157121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/05/2024] [Indexed: 04/25/2024] Open
Abstract
Affective touch-a slow, gentle, and pleasant form of touch-activates a different neural network than which is activated during discriminative touch in humans. Affective touch perception is enabled by specialized low-threshold mechanoreceptors in the skin with unmyelinated fibers called C tactile (CT) afferents. These CT afferents are conserved across mammalian species, including macaque monkeys. However, it is unknown whether the neural representation of affective touch is the same across species and whether affective touch's capacity to activate the hubs of the brain that compute socioaffective information requires conscious perception. Here, we used functional MRI to assess the preferential activation of neural hubs by slow (affective) vs. fast (discriminative) touch in anesthetized rhesus monkeys (Macaca mulatta). The insula, anterior cingulate cortex (ACC), amygdala, and secondary somatosensory cortex were all significantly more active during slow touch relative to fast touch, suggesting homologous activation of the interoceptive-allostatic network across primate species during affective touch. Further, we found that neural responses to affective vs. discriminative touch in the insula and ACC (the primary cortical hubs for interoceptive processing) changed significantly with age. Insula and ACC in younger animals differentiated between slow and fast touch, while activity was comparable between conditions for aged monkeys (equivalent to >70 y in humans). These results, together with prior studies establishing conserved peripheral nervous system mechanisms of affective touch transduction, suggest that neural responses to affective touch are evolutionarily conserved in monkeys, significantly impacted in old age, and do not necessitate conscious experience of touch.
Collapse
Affiliation(s)
- Joey A. Charbonneau
- Neuroscience Graduate Program, University of California, Davis, CA95616
- Neuroscience and Behavior Unit, California National Primate Research Center, University of California, Davis, CA95616
| | - Anthony C. Santistevan
- Neuroscience and Behavior Unit, California National Primate Research Center, University of California, Davis, CA95616
- Department of Psychology, University of California, Davis, CA95616
| | - Erika P. Raven
- Department of Radiology, Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY10016
| | - Jeffrey L. Bennett
- Neuroscience and Behavior Unit, California National Primate Research Center, University of California, Davis, CA95616
- Department of Psychology, University of California, Davis, CA95616
- Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, Sacramento, CA95817
- The Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Sacramento, CA95817
| | - Brian E. Russ
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY10962
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Department of Psychiatry, New York University Langone, New York, NY10016
| | - Eliza Bliss-Moreau
- Neuroscience and Behavior Unit, California National Primate Research Center, University of California, Davis, CA95616
- Department of Psychology, University of California, Davis, CA95616
| |
Collapse
|
29
|
Rosado MRS, Marzan-Rivera N, Watowich MM, Valle ADND, Pantoja P, Pavez-Fox MA, Siracusa ER, Cooper EB, Valle JEND, Phillips D, Ruiz-Lambides A, Martinez MI, Montague MJ, Platt ML, Higham JP, Brent LJN, Sariol CA, Snyder-Mackler N. Immune cell composition varies by age, sex and exposure to social adversity in free-ranging Rhesus Macaques. GeroScience 2024; 46:2107-2122. [PMID: 37853187 PMCID: PMC10828448 DOI: 10.1007/s11357-023-00962-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023] Open
Abstract
Increasing age is associated with dysregulated immune function and increased inflammation-patterns that are also observed in individuals exposed to chronic social adversity. Yet we still know little about how social adversity impacts the immune system and how it might promote age-related diseases. Here, we investigated how immune cell diversity varied with age, sex and social adversity (operationalized as low social status) in free-ranging rhesus macaques. We found age-related signatures of immunosenescence, including lower proportions of CD20 + B cells, CD20 + /CD3 + ratio, and CD4 + /CD8 + T cell ratio - all signs of diminished antibody production. Age was associated with higher proportions of CD3 + /CD8 + Cytotoxic T cells, CD16 + /CD3- Natural Killer cells, CD3 + /CD4 + /CD25 + and CD3 + /CD8 + /CD25 + T cells, and CD14 + /CD16 + /HLA-DR + intermediate monocytes, and lower levels of CD14 + /CD16-/HLA-DR + classical monocytes, indicating greater amounts of inflammation and immune dysregulation. We also found a sex-dependent effect of exposure to social adversity (i.e., low social status). High-status males, relative to females, had higher CD20 + /CD3 + ratios and CD16 + /CD3 Natural Killer cell proportions, and lower proportions of CD8 + Cytotoxic T cells. Further, low-status females had higher proportions of cytotoxic T cells than high-status females, while the opposite was observed in males. High-status males had higher CD20 + /CD3 + ratios than low-status males. Together, our study identifies the strong age and sex-dependent effects of social adversity on immune cell proportions in a human-relevant primate model. Thus, these results provide novel insights into the combined effects of demography and social adversity on immunity and their potential contribution to age-related diseases in humans and other animals.
Collapse
Affiliation(s)
- Mitchell R Sanchez Rosado
- Department of Microbiology & Medical Zoology, University of Puerto Rico-Medical Sciences, San Juan, PR, USA.
| | - Nicole Marzan-Rivera
- Department of Microbiology & Medical Zoology, University of Puerto Rico-Medical Sciences, San Juan, PR, USA
| | - Marina M Watowich
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | - Petraleigh Pantoja
- Department of Microbiology & Medical Zoology, University of Puerto Rico-Medical Sciences, San Juan, PR, USA
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Melissa A Pavez-Fox
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, EX4 4QG, UK
| | - Erin R Siracusa
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, EX4 4QG, UK
| | - Eve B Cooper
- Department of Anthropology, New York University, New York, NY, USA
| | - Josue E Negron-Del Valle
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Daniel Phillips
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Angelina Ruiz-Lambides
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Melween I Martinez
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Marketing, Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - James P Higham
- Department of Anthropology, New York University, New York, NY, USA
| | - Lauren J N Brent
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, EX4 4QG, UK
| | - Carlos A Sariol
- Department of Microbiology & Medical Zoology, University of Puerto Rico-Medical Sciences, San Juan, PR, USA
- Unit of Comparative Medicine, Caribbean Primate Research Center and Animal Resources Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Noah Snyder-Mackler
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA.
- School for Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
30
|
Bhatt LK, Shah CR, Patel SD, Patel SR, Patel VA, Patel RJ, Joshi NM, Shah NA, Patel JH, Dwivedi P, Sundar R, Jain MR. A Retrospective Comparison of Electrocardiographic Parameters in Ketamine and Tiletamine-Zolazepam Anesthetized Indian Rhesus Monkeys ( Macaca mulatta). Int J Toxicol 2024; 43:184-195. [PMID: 38108647 DOI: 10.1177/10915818231221276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Electrocardiographic evaluation is performed in rhesus monkeys to establish the cardiovascular safety of candidate molecules before progressing to clinical trials. These animals are usually immobilized chemically by ketamine (KTM) and tiletamine-zolazepam (TZ) to obtain a steady-state heart rate and to ensure adequate human safety. The present study aimed to evaluate the effect of these anesthetic regimens on different electrocardiographic parameters. Statistically significant lower HR and higher P-wave duration, RR, QRS, and QT intervals were observed in the KTM-anesthetized group in comparison to TZ-anesthetized animals. No significant changes were noticed in the PR interval and p-wave amplitude. Sex-based significance amongst these parameters was observed in male and female animals of TZ- and KTM-anesthetized groups. Regression analysis of four QTc formulas in TZ-anesthetized rhesus monkeys revealed that QTcNAK (Nakayama) better corrected the QT interval than QTcHAS (Hassimoto), QTcBZT (Bazett), and QTcFRD (Fridericia) formulas. QTcNAK exhibited the least correlation with the RR interval (slope closest to zero and r = .01) and displayed no statistical significance between male and female animals. These data will prove useful in the selection of anesthetic regimens for chemical restraint of rhesus monkeys in nonclinical safety evaluation studies.
Collapse
|
31
|
Smedley J. Editorial: Preclinical macaque models of viral diseases. Front Immunol 2023; 14:1331774. [PMID: 38022655 PMCID: PMC10666555 DOI: 10.3389/fimmu.2023.1331774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023] Open
Affiliation(s)
- Jeremy Smedley
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| |
Collapse
|
32
|
Newman LE, Testard C, DeCasien AR, Chiou KL, Watowich MM, Janiak MC, Pavez-Fox MA, Sanchez Rosado MR, Cooper EB, Costa CE, Petersen RM, Montague MJ, Platt ML, Brent LJN, Snyder-Mackler N, Higham JP. The biology of aging in a social world: Insights from free-ranging rhesus macaques. Neurosci Biobehav Rev 2023; 154:105424. [PMID: 37827475 PMCID: PMC10872885 DOI: 10.1016/j.neubiorev.2023.105424] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Social adversity can increase the age-associated risk of disease and death, yet the biological mechanisms that link social adversities to aging remain poorly understood. Long-term naturalistic studies of nonhuman animals are crucial for integrating observations of social behavior throughout an individual's life with detailed anatomical, physiological, and molecular measurements. Here, we synthesize the body of research from one such naturalistic study system, Cayo Santiago, which is home to the world's longest continuously monitored free-ranging population of rhesus macaques (Macaca mulatta). We review recent studies of age-related variation in morphology, gene regulation, microbiome composition, and immune function. We also discuss ecological and social modifiers of age-markers in this population. In particular, we summarize how a major natural disaster, Hurricane Maria, affected rhesus macaque physiology and social structure and highlight the context-dependent and domain-specific nature of aging modifiers. Finally, we conclude by providing directions for future study, on Cayo Santiago and elsewhere, that will further our understanding of aging across different domains and how social adversity modifies aging processes.
Collapse
Affiliation(s)
- Laura E Newman
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA.
| | - Camille Testard
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA.
| | - Alex R DeCasien
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Kenneth L Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Marina M Watowich
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA; Department of Biology, University of Washington, Seattle, WA, USA
| | - Mareike C Janiak
- Department of Anthropology, New York University, New York, NY, USA
| | - Melissa A Pavez-Fox
- Centre for Research in Animal Behaviour, University of Exeter, United Kingdom
| | | | - Eve B Cooper
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA
| | - Christina E Costa
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA
| | - Rachel M Petersen
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA; Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren J N Brent
- Centre for Research in Animal Behaviour, University of Exeter, United Kingdom
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - James P Higham
- Department of Anthropology, New York University, New York, NY, USA; New York Consortium in Evolutionary Primatology, New York, NY, USA
| |
Collapse
|
33
|
Rothwell ES, Carp SB, Bliss-Moreau E. The importance of social behavior in nonhuman primate studies of aging: A mini-review. Neurosci Biobehav Rev 2023; 154:105422. [PMID: 37806369 PMCID: PMC10716830 DOI: 10.1016/j.neubiorev.2023.105422] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/30/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Social behavior plays an important role in supporting both psychological and physical health across the lifespan. People's social lives change as they age, and the nature of these changes differ based on whether people are on healthy aging trajectories or are experiencing neurodegenerative diseases that cause dementia, such as Alzheimer's disease and Parkinson's disease. Nonhuman primate models of aging have provided a base of knowledge comparing aging trajectories in health and disease, but these studies rarely emphasize social behavior changes as a consequence of the aging process. What data exist hold particular value, as negative effects of disease and aging on social behavior are likely to have disproportionate impacts on quality of life. In this mini review, we examine the literature on nonhuman primate models of aging with a focus on social behavior, in the context of both health and disease. We propose that adopting a greater focus on social behavior outcomes in nonhuman primates will improve our understanding of the intersection of health, aging and sociality in humans.
Collapse
Affiliation(s)
- Emily S Rothwell
- Department of Neurobiology, School of Medicine University of Pittsburgh, 3501 Fifth Avenue, Biomedical Science Tower 3, Pittsburgh, PA 15213, USA.
| | - Sarah B Carp
- Neuroscience & Behavior Unit, California National Primate Research Center, University of California Davis, County Road 98 at Hutchinson Drive, Davis, CA 95616, USA
| | - Eliza Bliss-Moreau
- Neuroscience & Behavior Unit, California National Primate Research Center, University of California Davis, County Road 98 at Hutchinson Drive, Davis, CA 95616, USA; Department of Psychology, University of California Davis, County Road 98 at Hutchinson Drive, Davis, CA 95616, USA
| |
Collapse
|
34
|
Park EG, Lee YJ, Huh JW, Park SJ, Imai H, Kim WR, Lee DH, Kim JM, Shin HJ, Kim HS. Identification of microRNAs Derived from Transposable Elements in the Macaca mulatta (Rhesus Monkey) Genome. Genes (Basel) 2023; 14:1984. [PMID: 38002927 PMCID: PMC10671384 DOI: 10.3390/genes14111984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Transposable elements (TEs) are mobile DNA entities that can move within the host genome. Over long periods of evolutionary time, TEs are typically silenced via the accumulation of mutations in the genome, ultimately resulting in their immobilization. However, they still play an important role in the host genome by acting as regulatory elements. They influence host transcription in various ways, one of which as the origin of the generation of microRNAs (miRNAs), which are so-called miRNAs derived from TEs (MDTEs). miRNAs are small non-coding RNAs that are involved in many biological processes by regulating gene expression at the post-transcriptional level. Here, we identified MDTEs in the Macaca mulatta (rhesus monkey) genome, which is phylogenetically close species to humans, based on the genome coordinates of miRNAs and TEs. The expression of 5 out of 17 MDTEs that were exclusively registered in M. mulatta from the miRBase database (v22) was examined via quantitative polymerase chain reaction (qPCR). Moreover, Gene Ontology analysis was performed to examine the functional implications of the putative target genes of the five MDTEs.
Collapse
Affiliation(s)
- Eun Gyung Park
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (E.G.P.); (Y.J.L.); (W.R.K.); (D.H.L.); (J.-m.K.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Yun Ju Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (E.G.P.); (Y.J.L.); (W.R.K.); (D.H.L.); (J.-m.K.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (J.-W.H.); (S.-J.P.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Sang-Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (J.-W.H.); (S.-J.P.)
| | - Hiroo Imai
- Molecular Biology Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi 484-8506, Japan;
| | - Woo Ryung Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (E.G.P.); (Y.J.L.); (W.R.K.); (D.H.L.); (J.-m.K.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Du Hyeong Lee
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (E.G.P.); (Y.J.L.); (W.R.K.); (D.H.L.); (J.-m.K.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Jung-min Kim
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (E.G.P.); (Y.J.L.); (W.R.K.); (D.H.L.); (J.-m.K.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Hae Jin Shin
- Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea; (E.G.P.); (Y.J.L.); (W.R.K.); (D.H.L.); (J.-m.K.)
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Heui-Soo Kim
- Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
35
|
Chiou KL, Huang X, Bohlen MO, Tremblay S, DeCasien AR, O’Day DR, Spurrell CH, Gogate AA, Zintel TM, Andrews MG, Martínez MI, Starita LM, Montague MJ, Platt ML, Shendure J, Snyder-Mackler N. A single-cell multi-omic atlas spanning the adult rhesus macaque brain. SCIENCE ADVANCES 2023; 9:eadh1914. [PMID: 37824616 PMCID: PMC10569716 DOI: 10.1126/sciadv.adh1914] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Cataloging the diverse cellular architecture of the primate brain is crucial for understanding cognition, behavior, and disease in humans. Here, we generated a brain-wide single-cell multimodal molecular atlas of the rhesus macaque brain. Together, we profiled 2.58 M transcriptomes and 1.59 M epigenomes from single nuclei sampled from 30 regions across the adult brain. Cell composition differed extensively across the brain, revealing cellular signatures of region-specific functions. We also identified 1.19 M candidate regulatory elements, many previously unidentified, allowing us to explore the landscape of cis-regulatory grammar and neurological disease risk in a cell type-specific manner. Altogether, this multi-omic atlas provides an open resource for investigating the evolution of the human brain and identifying novel targets for disease interventions.
Collapse
Affiliation(s)
- Kenneth L. Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Xingfan Huang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Martin O. Bohlen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sébastien Tremblay
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex R. DeCasien
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Diana R. O’Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Cailyn H. Spurrell
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Aishwarya A. Gogate
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Trisha M. Zintel
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Cayo Biobank Research Unit
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Caribbean Primate Research Center, University of Puerto Rico, San Juan, PR, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Madeline G. Andrews
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Melween I. Martínez
- Caribbean Primate Research Center, University of Puerto Rico, San Juan, PR, USA
| | - Lea M. Starita
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Michael J. Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
36
|
Berendzen KM, Bales KL, Manoli DS. Attachment across the lifespan: Examining the intersection of pair bonding neurobiology and healthy aging. Neurosci Biobehav Rev 2023; 153:105339. [PMID: 37536581 PMCID: PMC11073483 DOI: 10.1016/j.neubiorev.2023.105339] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/17/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Increasing evidence suggests that intact social bonds are protective against age-related morbidity, while bond disruption and social isolation increase the risk for multiple age-related diseases. Social attachments, the enduring, selective bonds formed between individuals, are thus essential to human health. Socially monogamous species like the prairie vole (M. ochrogaster) form long-term pair bonds, allowing us to investigate the mechanisms underlying attachment and the poorly understood connection between social bonds and health. In this review, we explore several potential areas of focus emerging from data in humans and other species associating attachment and healthy aging, and evidence from prairie voles that may clarify this link. We examine gaps in our understanding of social cognition and pair bond behavior. Finally, we discuss physiologic pathways related to pair bonding that promote resilience to the processes of aging and age-related disease. Advances in the development of molecular genetic tools in monogamous species will allow us to bridge the mechanistic gaps presented and identify conserved research and therapeutic targets relevant to human health and aging.
Collapse
Affiliation(s)
- Kristen M. Berendzen
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco; San Francisco, CA 95158, USA
- Center for Integrative Neuroscience, University of California, San Francisco; San Francisco, CA 95158, USA
- Weill Institute for Neurosciences, University of California, San Francisco; San Francisco, CA 95158, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco; San Francisco, CA 95158, USA
| | - Karen L. Bales
- Department of Psychology, University of California, Davis; Davis, CA 95616, USA
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis; Davis, CA 95616, USA
| | - Devanand S. Manoli
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco; San Francisco, CA 95158, USA
- Center for Integrative Neuroscience, University of California, San Francisco; San Francisco, CA 95158, USA
- Weill Institute for Neurosciences, University of California, San Francisco; San Francisco, CA 95158, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco; San Francisco, CA 95158, USA
- Neurosciences Graduate Program, University of California, San Francisco; San Francisco, CA 95158, USA
| |
Collapse
|
37
|
Patterson SK, Petersen RM, Brent LJN, Snyder-Mackler N, Lea AJ, Higham JP. Natural Animal Populations as Model Systems for Understanding Early Life Adversity Effects on Aging. Integr Comp Biol 2023; 63:681-692. [PMID: 37279895 PMCID: PMC10503476 DOI: 10.1093/icb/icad058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 05/30/2023] [Indexed: 06/08/2023] Open
Abstract
Adverse experiences in early life are associated with aging-related disease risk and mortality across many species. In humans, confounding factors, as well as the difficulty of directly measuring experiences and outcomes from birth till death, make it challenging to identify how early life adversity impacts aging and health. These challenges can be mitigated, in part, through the study of non-human animals, which are exposed to parallel forms of adversity and can age similarly to humans. Furthermore, studying the links between early life adversity and aging in natural populations of non-human animals provides an excellent opportunity to better understand the social and ecological pressures that shaped the evolution of early life sensitivities. Here, we highlight ongoing and future research directions that we believe will most effectively contribute to our understanding of the evolution of early life sensitivities and their repercussions.
Collapse
Affiliation(s)
- Sam K Patterson
- Department of Anthropology, New York University, New York City, 10003, USA
| | - Rachel M Petersen
- Department of Biological Science, Vanderbilt University, Nashville, 37232, USA
| | - Lauren J N Brent
- Department of Psychology, University of Exeter, Exeter, EX4 4QG, United Kingdom
| | - Noah Snyder-Mackler
- School of Life Sciences, Center for Evolution and Medicine, and School of Human Evolution and Social Change, Arizona State University, Tempe, 85281, USA
| | - Amanda J Lea
- Department of Biological Science, Vanderbilt University, Nashville, 37232, USA
- Child and Brain Development Program, Canadian Institute for Advanced Study, Toronto, M5G 1M1, Canada
| | - James P Higham
- Department of Anthropology, New York University, New York City, 10003, USA
| |
Collapse
|
38
|
Pittet F, Hinde K. Meager Milk: Lasting Consequences for Adult Daughters of Primiparous Mothers Among Rhesus Macaques (Macaca mulatta). Integr Comp Biol 2023; 63:569-584. [PMID: 37170073 PMCID: PMC10503474 DOI: 10.1093/icb/icad022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023] Open
Abstract
Among mammals, primipara who initiate reproduction before full maturity can be constrained in their maternal investment, both due to fewer somatic resources and tradeoffs between their own continued development and reproductive effort. Primipara are particularly limited in their capacity to synthesize milk during lactation, the costliest aspect of reproduction for most mammals, especially primates due to long periods of postnatal development. Due to reduced milk transfer, Firstborns may be at elevated risk for long-term consequences of deficits in early life endowment from their primiparous mothers. Here we investigated mass, growth, stature, and lactation performance among N = 273 adult daughters across N = 335 reproductions, who were their own mother's Firstborn or Laterborn progeny, among rhesus macaques (Macaca mulatta) at the California National Primate Research Center. We further explored mass during infancy of the offspring of Firstborn and Laterborn mothers. Firstborns had accelerated growth during infancy, but had slowed growth during juvenility, compared to Laterborns. Although both Firstborns and Laterborns were the same age at reproductive debut, Firstborns had lower body mass, an effect that persisted throughout the reproductive career. Available milk energy, the product of milk energetic density and milk yield, was on average 16% lower for Firstborns compared to Laterborns, a difference that was only partially mediated by their lower mass. Despite differences in their mothers' energy provision through milk, the mass of infants of Firstborn and Laterborn mothers did not differ at peak lactation, suggesting that infants of Firstborns devote a higher proportion of milk energy to growth than infants of Laterborns. To date few studies have explored how early life conditions shape capacities to synthesize milk and milk composition. Our findings contribute new information among primates on how early life maternal endowments are associated with persistent effects long after the period of maternal dependence well into reproductive maturity.
Collapse
Affiliation(s)
- Florent Pittet
- Neuroscience and Behavior Unit, California National Primate Research Center, University of California, Davis, CA 95616, USA
| | - Katie Hinde
- School of Human Evolution and Social Change, Tempe, AZ 85287, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
39
|
Sazhnyev Y, Sin TN, Ma A, Chang E, Huynh L, Roszak K, Park S, Choy K, Farsiu S, Moshiri A, Thomasy SM, Yiu G. Choroidal Changes in Rhesus Macaques in Aging and Age-Related Drusen. Invest Ophthalmol Vis Sci 2023; 64:44. [PMID: 37773500 PMCID: PMC10547013 DOI: 10.1167/iovs.64.12.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/07/2023] [Indexed: 10/01/2023] Open
Abstract
Purpose Choroidal vascular changes occur with normal aging and age-related macular degeneration (AMD). Here, we evaluate choroidal thickness and vascularity in aged rhesus macaques to better understand the choroid's role in this nonhuman primate model of AMD. Methods We analyzed optical coherence tomography (OCT) images of 244 eyes from 122 rhesus macaques (aged 4-32 years) to measure choroidal thickness (CT) and choroidal vascularity index (CVI). Drusen number, size, and volume were measured by semiautomated annotation and segmentation of OCT images. We performed regression analyses to determine any association of CT or CVI with age, sex, and axial length and to determine if the presence and volume of soft drusen impacted these choroidal parameters. Results In rhesus macaques, subfoveal CT decreased with age at 3.2 µm/y (R2 = 0.481, P < 0.001), while CVI decreased at 0.66% per year (R2 = 0.257, P < 0.001). Eyes with soft drusen exhibited thicker choroid (179.9 ± 17.5 µm vs. 162.0 ± 27.9 µm, P < 0.001) and higher CVI (0.612 ± 0.051 vs. 0.577 ± 0.093, P = 0.005) than age-matched control animals. Neither CT or CVI appeared to be associated with drusen number, size, or volume in this cohort. However, some drusen in macaques were associated with underlying choroidal vessel enlargement resembling pachydrusen in human patients with AMD. Conclusions Changes in the choroidal vasculature in rhesus macaques resemble choroidal changes in human aging, but eyes with drusen exhibit choroidal thickening, increased vascularity, and phenotypic characteristics of pachydrusen observed in some patients with AMD.
Collapse
Affiliation(s)
- Yevgeniy Sazhnyev
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
- Department of Ophthalmology, California Northstate University, College of Medicine, Elk Grove, California, United States
| | - Tzu-Ni Sin
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Anthony Ma
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
- Department of Ophthalmology, California Northstate University, College of Medicine, Elk Grove, California, United States
| | - Ellie Chang
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Leon Huynh
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Karolina Roszak
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Sangwan Park
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Kevin Choy
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Sina Farsiu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Sara M. Thomasy
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| |
Collapse
|
40
|
Thompson González N, Machanda Z, Emery Thompson M. Age-related social selectivity: An adaptive lens on a later life social phenotype. Neurosci Biobehav Rev 2023; 152:105294. [PMID: 37380041 PMCID: PMC10529433 DOI: 10.1016/j.neubiorev.2023.105294] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Age-related social selectivity is a process in which older humans reduce their number of social partners to a subset of positive and emotionally fulfilling relationships. Although selectivity has been attributed to humans' unique perceptions of time horizons, recent evidence demonstrates that these social patterns and processes occur in other non-human primates, suggesting an evolutionarily wider phenomenon. Here, we develop the hypothesis that selective social behavior is an adaptive strategy that allows social animals to balance the costs and benefits of navigating social environments in the face of age-related functional declines. We first aim to distinguish social selectivity from the non-adaptive social consequences of aging. We then outline multiple mechanisms by which social selectivity in old age may enhance fitness and healthspan. Our goal is to lay out a research agenda to identify selective strategies and their potential benefits. Given the importance of social support for health across primates, understanding why aging individuals lose social connections and how they can remain resilient has vital applications to public health research.
Collapse
Affiliation(s)
- Nicole Thompson González
- Integrative Anthropological Sciences, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Zarin Machanda
- Department of Anthropology, Tufts University, Medford, MA 02155, USA
| | | |
Collapse
|
41
|
Datta D. Interrogating the Etiology of Sporadic Alzheimer's Disease Using Aging Rhesus Macaques: Cellular, Molecular, and Cortical Circuitry Perspectives. J Gerontol A Biol Sci Med Sci 2023; 78:1523-1534. [PMID: 37279946 PMCID: PMC10460555 DOI: 10.1093/gerona/glad134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Indexed: 06/08/2023] Open
Abstract
Aging is the most significant risk factor for neurodegenerative disorders such as Alzheimer's disease (AD) associated with profound socioeconomic and personal costs. Consequently, there is an urgent need for animal models that recapitulate the age-related spatial and temporal complexity and patterns of pathology identical to human AD. Our research in aging nonhuman primate models involving rhesus macaques has revealed naturally occurring amyloid and tau pathology, including the formation of amyloid plaques and neurofibrillary tangles comprising hyperphosphorylated tau. Moreover, rhesus macaques exhibit synaptic dysfunction in association cortices and cognitive impairments with advancing age, and thus can be used to interrogate the etiological mechanisms that generate neuropathological cascades in sporadic AD. Particularly, unique molecular mechanisms (eg, feedforward cyclic adenosine 3',5'-monophosphate [cAMP]-Protein kinase A (PKA)-calcium signaling) in the newly evolved primate dorsolateral prefrontal cortex are critical for persistent firing required for subserving higher-order cognition. For example, dendritic spines in primate dorsolateral prefrontal cortex contain a specialized repertoire of proteins to magnify feedforward cAMP-PKA-calcium signaling such as N-methyl-d-aspartic acid receptors and calcium channels on the smooth endoplasmic reticulum (eg, ryanodine receptors). This process is constrained by phosphodiesterases (eg, PDE4) that hydrolyze cAMP and calcium-buffering proteins (eg, calbindin) in the cytosol. However, genetic predispositions and age-related insults exacerbate feedforward cAMP-Protein kinase A-calcium signaling pathways that induce a myriad of downstream effects, including the opening of K+ channels to weaken network connectivity, calcium-mediated dysregulation of mitochondria, and activation of inflammatory cascades to eliminate synapses, thereby increasing susceptibility to atrophy. Therefore, aging rhesus macaques provide an invaluable model to explore novel therapeutic strategies in sporadic AD.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
42
|
Jagadesan S, Mondal P, Carlson MA, Guda C. Evaluation of Five Mammalian Models for Human Disease Research Using Genomic and Bioinformatic Approaches. Biomedicines 2023; 11:2197. [PMID: 37626695 PMCID: PMC10452283 DOI: 10.3390/biomedicines11082197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
The suitability of an animal model for use in studying human diseases relies heavily on the similarities between the two species at the genetic, epigenetic, and metabolic levels. However, there is a lack of consistent data from different animal models at each level to evaluate this suitability. With the availability of genome sequences for many mammalian species, it is now possible to compare animal models based on genomic similarities. Herein, we compare the coding sequences (CDSs) of five mammalian models, including rhesus macaque, marmoset, pig, mouse, and rat models, with human coding sequences. We identified 10,316 conserved CDSs across the five organisms and the human genome based on sequence similarity. Mapping the human-disease-associated single-nucleotide polymorphisms (SNPs) from these conserved CDSs in each species has identified species-specific associations with various human diseases. While associations with a disease such as colon cancer were prevalent in multiple model species, the rhesus macaque showed the most model-specific human disease associations. Based on the percentage of disease-associated SNP-containing genes, marmoset models are well suited to study many human ailments, including behavioral and cardiovascular diseases. This study demonstrates a genomic similarity evaluation of five animal models against human CDSs that could help investigators select a suitable animal model for studying their target disease.
Collapse
Affiliation(s)
- Sankarasubramanian Jagadesan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.J.); (M.A.C.)
| | - Pinaki Mondal
- Department of Surgery and Center for Advanced Surgical Technology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Mark A. Carlson
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.J.); (M.A.C.)
- Department of Surgery and Center for Advanced Surgical Technology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.J.); (M.A.C.)
- Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
43
|
Higham JP, Cooper EB, Whalen C, Stahl-Hennig C, Giavedoni LD, Heistermann M. Urinary cytokine measurements do not reflect surgery-induced inflammation in rhesus macaques. Am J Primatol 2023; 85:e23506. [PMID: 37222418 DOI: 10.1002/ajp.23506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 03/14/2023] [Accepted: 04/29/2023] [Indexed: 05/25/2023]
Abstract
Measurement of the health and disease status of free-ranging primates is often limited by a lack of available biomarkers of immune activation and inflammation that can be applied noninvasively via the measurement of urine or fecal samples. Here, we evaluate the potential usefulness of noninvasive urinary measurements of a number of cytokines, chemokines, and other markers of inflammation and infection. We took advantage of surgery-associated inflammation in seven captive rhesus macaques, collecting urine samples before and after the medical interventions. We measured these urine samples for 33 different markers of inflammation and immune activation that are known to be responsive to inflammation and infection in rhesus macaque blood samples, via the Luminex platform. We also measured all samples for concentrations of the soluble urokinase plasminogen activator receptor (suPAR), which we had validated in a prior study as an effective biomarker of inflammation. Despite urine samples being collected in captivity under ideal conditions (clean, no contamination with feces or soil, frozen quickly), 13/33 biomarkers measured via Luminex were found at concentrations below detection limits in >50% of samples. Of the remaining 20 markers, only 2 showed significant increases in response to surgery-IL18 and MPO (myeloperoxidase). However, suPAR measurements of the same samples show a consistent marked increase in response to surgery that is absent from the patterns of IL18 and MPO measurement. Given that our samples were collected under conditions that are greatly preferable to those usually encountered in the field, urinary cytokine measurements via the Luminex platform seem overall unpromising for primate field studies.
Collapse
Affiliation(s)
- James P Higham
- Department of Anthropology, New York University, New York, New York, USA
| | - Eve B Cooper
- Department of Anthropology, New York University, New York, New York, USA
| | - Connor Whalen
- Department of Anthropology, New York University, New York, New York, USA
| | | | - Luis D Giavedoni
- Southwest National Primate Research Center, Texas Biomedical Research Institute, Texas, San Antonio, USA
- Department of Biology, Trinity University, San Antonio, Texas, USA
| | | |
Collapse
|
44
|
Santus L, Sopena-Rios M, García-Pérez R, Lin AE, Adams GC, Barnes KG, Siddle KJ, Wohl S, Reverter F, Rinn JL, Bennett RS, Hensley LE, Sabeti PC, Melé M. Single-cell profiling of lncRNA expression during Ebola virus infection in rhesus macaques. Nat Commun 2023; 14:3866. [PMID: 37391481 PMCID: PMC10313701 DOI: 10.1038/s41467-023-39627-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/19/2023] [Indexed: 07/02/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are involved in numerous biological processes and are pivotal mediators of the immune response, yet little is known about their properties at the single-cell level. Here, we generate a multi-tissue bulk RNAseq dataset from Ebola virus (EBOV) infected and not-infected rhesus macaques and identified 3979 novel lncRNAs. To profile lncRNA expression dynamics in immune circulating single-cells during EBOV infection, we design a metric, Upsilon, to estimate cell-type specificity. Our analysis reveals that lncRNAs are expressed in fewer cells than protein-coding genes, but they are not expressed at lower levels nor are they more cell-type specific when expressed in the same number of cells. In addition, we observe that lncRNAs exhibit similar changes in expression patterns to those of protein-coding genes during EBOV infection, and are often co-expressed with known immune regulators. A few lncRNAs change expression specifically upon EBOV entry in the cell. This study sheds light on the differential features of lncRNAs and protein-coding genes and paves the way for future single-cell lncRNA studies.
Collapse
Affiliation(s)
- Luisa Santus
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia, 08034, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Maria Sopena-Rios
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia, 08034, Spain
| | - Raquel García-Pérez
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia, 08034, Spain
| | - Aaron E Lin
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Program in Virology, Harvard Medical School, Boston, MA, 02115, USA
| | - Gordon C Adams
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Kayla G Barnes
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
- Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Katherine J Siddle
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Shirlee Wohl
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- The Scripps Research Institute, Department of Immunology and Microbiology, La Jolla, CA, USA
| | - Ferran Reverter
- Department of Genetics, Microbiology and Statistics University of Barcelona, Barcelona, Spain
| | - John L Rinn
- Department of Biochemistry, University of Colorado Boulder, Boulder, 80303, USA
| | - Richard S Bennett
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Lisa E Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA.
| | - Pardis C Sabeti
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Harvard Program in Virology, Harvard Medical School, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA.
| | - Marta Melé
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia, 08034, Spain.
| |
Collapse
|
45
|
Hendrickson SM, Thomas A, Raué HP, Prongay K, Haertel AJ, Rhoades NS, Slifka JF, Gao L, Quintel BK, Amanna IJ, Messaoudi I, Slifka MK. Campylobacter vaccination reduces diarrheal disease and infant growth stunting among rhesus macaques. Nat Commun 2023; 14:3806. [PMID: 37365162 PMCID: PMC10293212 DOI: 10.1038/s41467-023-39433-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Campylobacter-associated enteric disease is estimated to be responsible for more than 160 million cases of gastroenteritis each year and is linked to growth stunting of infants living under conditions of poor sanitation and hygiene. Here, we examine naturally occurring Campylobacter-associated diarrhea among rhesus macaques as a model to determine if vaccination could reduce severe diarrheal disease and infant growth stunting. Compared to unvaccinated controls, there are no Campylobacter diarrhea-associated deaths observed among vaccinated infant macaques and all-cause diarrhea-associated infant mortality is decreased by 76% (P = 0.03). By 9 months of age, there is a 1.3 cm increase in dorsal length that equaled a significant 1.28 LAZ (Length-for-Age Z score) improvement in linear growth among vaccinated infants compared to their unvaccinated counterparts (P = 0.001). In this work, we show that Campylobacter vaccination not only reduces diarrheal disease but also potentially serves as an effective intervention that improves infant growth trajectories.
Collapse
Affiliation(s)
- Sara M Hendrickson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Archana Thomas
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Kamm Prongay
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Andrew J Haertel
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Nicholas S Rhoades
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY, 40506, USA
| | - Jacob F Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Lina Gao
- Biostatistics and Bioinformatics Core, Oregon National Primate Research Center, Biostatistics Shared Resource, Knight Cancer Institute, Portland, OR, 97239, USA
| | | | - Ian J Amanna
- Najít Technologies, Inc., Beaverton, OR, 97006, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY, 40506, USA
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA.
| |
Collapse
|
46
|
Siracusa ER, Pereira AS, Brask JB, Negron-Del Valle JE, Phillips D, Platt ML, Higham JP, Snyder-Mackler N, Brent LJN. Ageing in a collective: the impact of ageing individuals on social network structure. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220061. [PMID: 36802789 PMCID: PMC9939263 DOI: 10.1098/rstb.2022.0061] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/16/2022] [Indexed: 02/21/2023] Open
Abstract
Ageing affects many phenotypic traits, but its consequences for social behaviour have only recently become apparent. Social networks emerge from associations between individuals. The changes in sociality that occur as individuals get older are thus likely to impact network structure, yet this remains unstudied. Here we use empirical data from free-ranging rhesus macaques and an agent-based model to test how age-based changes in social behaviour feed up to influence: (i) an individual's level of indirect connectedness in their network and (ii) overall patterns of network structure. Our empirical analyses revealed that female macaques became less indirectly connected as they aged for some, but not for all network measures examined. This suggests that indirect connectivity is affected by ageing, and that ageing animals can remain well integrated in some social contexts. Surprisingly, we did not find evidence for a relationship between age distribution and the structure of female macaque networks. We used an agent-based model to gain further understanding of the link between age-based differences in sociality and global network structure, and under which circumstances global effects may be detectable. Overall, our results suggest a potentially important and underappreciated role of age in the structure and function of animal collectives, which warrants further investigation. This article is part of a discussion meeting issue 'Collective behaviour through time'.
Collapse
Affiliation(s)
- Erin R. Siracusa
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter EX4 4QG, UK
| | - André S. Pereira
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter EX4 4QG, UK
- Research Centre for Anthropology and Health, Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Josefine Bohr Brask
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark
| | | | - Daniel Phillips
- Center for Evolution and Medicine, Arizona State University, Arizona, AZ 85281, USA
| | - Cayo Biobank Research Unit
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter EX4 4QG, UK
- Research Centre for Anthropology and Health, Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, DK-2800, Kongens Lyngby, Denmark
- Center for Evolution and Medicine, Arizona State University, Arizona, AZ 85281, USA
- School of Life Sciences, Arizona State University, Arizona, AZ 85281, USA
- School for Human Evolution and Social Change, Arizona State University, Arizona, AZ 85281, USA
- Department of Neuroscience, University of Pennsylvania, PA 19104, USA
- Department of Psychology, University of Pennsylvania, PA 19104, USA
- Department of Marketing, University of Pennsylvania, PA 19104, USA
- Department of Anthropology, New York University, New York, NY 10003, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, PA 19104, USA
- Department of Psychology, University of Pennsylvania, PA 19104, USA
- Department of Marketing, University of Pennsylvania, PA 19104, USA
| | - James P. Higham
- Department of Anthropology, New York University, New York, NY 10003, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Arizona, AZ 85281, USA
- School of Life Sciences, Arizona State University, Arizona, AZ 85281, USA
- School for Human Evolution and Social Change, Arizona State University, Arizona, AZ 85281, USA
| | - Lauren J. N. Brent
- School of Psychology, Centre for Research in Animal Behaviour, University of Exeter, Exeter EX4 4QG, UK
| |
Collapse
|
47
|
Milligan EC, Olstad K, Williams CA, Mallory M, Cano P, Cross KA, Munt JE, Garrido C, Lindesmith L, Watanabe J, Usachenko JL, Hopkins L, Immareddy R, Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Sammak RL, Pollard RE, Yee JL, Herbek S, Scobey T, Miehlke D, Fouda G, Ferrari G, Gao H, Shen X, Kozlowski PA, Montefiori D, Hudgens MG, Edwards DK, Carfi A, Corbett KS, Graham BS, Fox CB, Tomai M, Iyer SS, Baric R, Reader R, Dittmer DP, Van Rompay KKA, Permar SR, De Paris K. Infant rhesus macaques immunized against SARS-CoV-2 are protected against heterologous virus challenge 1 year later. Sci Transl Med 2023; 15:eadd6383. [PMID: 36454813 PMCID: PMC9765459 DOI: 10.1126/scitranslmed.add6383] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The U.S. Food and Drug Administration only gave emergency use authorization of the BNT162b2 and mRNA-1273 SARS-CoV-2 vaccines for infants 6 months and older in June 2022. Yet questions regarding the durability of vaccine efficacy, especially against emerging variants, in this age group remain. We demonstrated previously that a two-dose regimen of stabilized prefusion Washington SARS-CoV-2 S-2P spike (S) protein encoded by mRNA encapsulated in lipid nanoparticles (mRNA-LNP) or purified S-2P mixed with 3M-052, a synthetic Toll-like receptor (TLR) 7/8 agonist, in a squalene emulsion (Protein+3M-052-SE) was safe and immunogenic in infant rhesus macaques. Here, we demonstrate that broadly neutralizing and spike-binding antibodies against variants of concern (VOCs), as well as T cell responses, persisted for 12 months. At 1 year, corresponding to human toddler age, we challenged vaccinated rhesus macaques and age-matched nonvaccinated controls intranasally and intratracheally with a high dose of heterologous SARS-CoV-2 B.1.617.2 (Delta). Seven of eight control rhesus macaques exhibited severe interstitial pneumonia and high virus replication in the upper and lower respiratory tract. In contrast, vaccinated rhesus macaques had faster viral clearance with mild to no pneumonia. Neutralizing and binding antibody responses to the B.1.617.2 variant at the day of challenge correlated with lung pathology and reduced virus replication. Overall, the Protein+3M-052-SE vaccine provided superior protection to the mRNA-LNP vaccine, emphasizing opportunities for optimization of current vaccine platforms. The observed efficacy of both vaccines 1 year after vaccination supports the implementation of an early-life SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Emma C Milligan
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine Olstad
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael Mallory
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patricio Cano
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaitlyn A Cross
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer E Munt
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carolina Garrido
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Lisa Lindesmith
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Jodie L Usachenko
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Lincoln Hopkins
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Ramya Immareddy
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | | | - Sonny R Elizaldi
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Rebecca L Sammak
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Rachel E Pollard
- School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, USA
| | - JoAnn L Yee
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Trevor Scobey
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dieter Miehlke
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Genevieve Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Hongmei Gao
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael G Hudgens
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | - Kizzmekia S Corbett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Christopher B Fox
- Access to Advanced Health Institute, Seattle, WA 98102, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Mark Tomai
- 3M Corporate Research Materials Laboratory, Saint Paul, MN 55144, USA
| | - Smita S Iyer
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Ralph Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel Reader
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Department of Pathology, Microbiology and Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
48
|
Wildenberg G, Li H, Kasthuri N. The Development of Synapses in Mouse and Macaque Primary Sensory Cortices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528564. [PMID: 36824798 PMCID: PMC9949058 DOI: 10.1101/2023.02.15.528564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
We report that the rate of synapse development in primary sensory cortices of mice and macaques is unrelated to lifespan, as was previously thought. We analyzed 28,084 synapses over multiple developmental time points in both species and find, instead, that net excitatory synapse development of mouse and macaque neurons primarily increased at similar rates in the first few postnatal months, and then decreased over a span of 1-1.5 years of age. The development of inhibitory synapses differed qualitatively across species. In macaques, net inhibitory synapses first increase and then decrease on excitatory soma at similar ages as excitatory synapses. In mice, however, such synapses are added throughout life. These findings contradict the long-held belief that the cycle of synapse formation and pruning occurs earlier in shorter-lived animals. Instead, our results suggest more nuanced rules, with the development of different types of synapses following different timing rules or different trajectories across species.
Collapse
Affiliation(s)
- Gregg Wildenberg
- Department of Neurobiology, The University of Chicago
- Argonne National Laboratory, Biosciences Division
| | - Hanyu Li
- Department of Neurobiology, The University of Chicago
- Argonne National Laboratory, Biosciences Division
| | - Narayanan Kasthuri
- Department of Neurobiology, The University of Chicago
- Argonne National Laboratory, Biosciences Division
| |
Collapse
|
49
|
Newman LE, Testard C, DeCasien AR, Chiou KL, Watowich MM, Janiak MC, Pavez-Fox MA, Rosado MRS, Cooper EB, Costa CE, Petersen RM, Montague MJ, Platt ML, Brent LJ, Snyder-Mackler N, Higham JP. The biology of aging in a social world:insights from free-ranging rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.28.525893. [PMID: 36747827 PMCID: PMC9900930 DOI: 10.1101/2023.01.28.525893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Social adversity can increase the age-associated risk of disease and death, yet the biological mechanisms that link social adversities to aging remain poorly understood. Long-term naturalistic studies of nonhuman animals are crucial for integrating observations of social behavior throughout an individual's life with detailed anatomical, physiological, and molecular measurements. Here, we synthesize the body of research from one such naturalistic study system, Cayo Santiago Island, which is home to the world's longest continuously monitored free-ranging population of rhesus macaques. We review recent studies of age-related variation in morphology, gene regulation, microbiome composition, and immune function. We also discuss ecological and social modifiers of age-markers in this population. In particular, we summarize how a major natural disaster, Hurricane Maria, affected rhesus macaque physiology and social structure and highlight the context-dependent and domain-specific nature of aging modifiers. Finally, we conclude by providing directions for future study, on Cayo Santiago and elsewhere, that will further our understanding of aging across different domains and how social adversity modifies aging processes.
Collapse
Affiliation(s)
- Laura E. Newman
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| | - Camille Testard
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex R. DeCasien
- Section on Developmental Neurogenomics, National Institutes of Mental Health, Bethesda, Maryland, USA
| | - Kenneth L. Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Marina M. Watowich
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Mareike C. Janiak
- Department of Anthropology, New York University, New York, New York, USA
| | | | | | - Eve B. Cooper
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| | - Christina E. Costa
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| | - Rachel M. Petersen
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Michael J. Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren J.N. Brent
- Centre for Research in Animal Behaviour, University of Exeter, United Kingdom
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - James P. Higham
- Department of Anthropology, New York University, New York, New York, USA
- The New York Consortium in Evolutionary Primatology (NYCEP), New York, New York, USA
| |
Collapse
|
50
|
Within-individual changes reveal increasing social selectivity with age in rhesus macaques. Proc Natl Acad Sci U S A 2022; 119:e2209180119. [PMID: 36445967 PMCID: PMC9894112 DOI: 10.1073/pnas.2209180119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Accumulating evidence in humans and other mammals suggests older individuals tend to have smaller social networks. Uncovering the cause of these declines can inform how changes in social relationships with age affect health and fitness in later life. While age-based declines in social networks have been thought to be detrimental, physical and physiological limitations associated with age may lead older individuals to adjust their social behavior and be more selective in partner choice. Greater selectivity with age has been shown in humans, but the extent to which this phenomenon occurs across the animal kingdom remains an open question. Using longitudinal data from a population of rhesus macaques on Cayo Santiago, we provide compelling evidence in a nonhuman animal for within-individual increases in social selectivity with age. Our analyses revealed that adult female macaques actively reduced the size of their networks as they aged and focused on partners previously linked to fitness benefits, including kin and partners to whom they were strongly and consistently connected earlier in life. Females spent similar amounts of time socializing as they aged, suggesting that network shrinkage does not result from lack of motivation or ability to engage, nor was this narrowing driven by the deaths of social partners. Furthermore, females remained attractive companions and were not isolated by withdrawal of social partners. Taken together, our results provide rare empirical evidence for social selectivity in nonhumans, suggesting that patterns of increasing selectivity with age may be deeply rooted in primate evolution.
Collapse
|