1
|
Luciano A, Churchill GA. The impact of co-housing on murine aging studies. GeroScience 2025:10.1007/s11357-024-01480-x. [PMID: 39806236 DOI: 10.1007/s11357-024-01480-x] [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: 10/07/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Analysis of preclinical lifespan studies often assume that outcome data from co-housed animals are independent. In practice, treatments, such as controlled feeding or putative life-extending compounds, are applied to whole housing units, and as a result, the outcomes are potentially correlated within housing units. We consider intra-class (here, intra-cage) correlation in three published and two unpublished lifespan studies of aged mice encompassing more than 20,000 observations. We show that the independence assumption underlying common analytic techniques does not hold in these data, particularly for traits associated with frailty. We describe and demonstrate various analytical tools available to accommodate this study design and highlight a limitation of standard variance components models (i.e., linear mixed models) which are the usual statistical tools for handling correlated errors. Through simulations, we examine the statistical biases resulting from intra-cage correlations with similar magnitudes as observed in these case studies and discuss implications for power and reproducibility.
Collapse
|
2
|
Beam TC, Bright M, Pearson AC, Dua I, Smith M, Dutta AK, Bhadra SC, Salman S, Strickler CN, Anderson CE, Peshkin L, Yampolsky LY. Short lifespan is one's fate, long lifespan is one's achievement: lessons from Daphnia. GeroScience 2024; 46:6361-6381. [PMID: 38900345 PMCID: PMC11493910 DOI: 10.1007/s11357-024-01244-7] [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: 04/30/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Studies of longevity rely on baseline life expectancy of reference genotypes measured in standardized conditions. Variation among labs, protocols, and genotypes makes longevity intervention studies difficult to compare. Furthermore, extending lifespan under suboptimal conditions or that of a short-lived genotype may be of a lesser theoretical and translational value than extending the maximal possible lifespan. Daphnia is becoming a model organism of choice for longevity research complementing data obtained on traditional models. In this study, we report longevity of several genotypes of a long-lived species D. magna under a variety of protocols, aiming to document the highest lifespan, factors reducing it, and parameters that change with age and correlate with longevity. Combining longevity data from 25 experiments across two labs, we report a strong intraspecific variation, moderate effects of group size and medium composition, and strong genotype-by-environment interactions with respect to food level. Specifically, short-lived genotypes show no caloric restriction (CR) effect, while long-lived ones expand their lifespan even further under CR. We find that the CR non-responsive clones show little correlation between longevity and two measures of lipid peroxidation. In contrast, the long-lived, CR-responsive clones show a positive correlation between longevity and lipid hydroperoxide abundance, and a negative correlation with MDA concentration. This indicates differences among genotypes in age-related accumulation and detoxification of LPO products and their effects on longevity. Our observations support the hypothesis that a long lifespan can be affected by CR and levels of oxidative damage, while genetically determined short lifespan remains short regardless.
Collapse
Affiliation(s)
- Thomas C Beam
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Mchale Bright
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Amelia C Pearson
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Ishaan Dua
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Meridith Smith
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Ashit K Dutta
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Shymal C Bhadra
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, 46805, USA
| | - Saad Salman
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Caleb N Strickler
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Cora E Anderson
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Leonid Peshkin
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Lev Y Yampolsky
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA.
| |
Collapse
|
3
|
Bystrykh LV. Why an integrated view of gene expression studies on hematopoiesis in mouse aging is better than the sum of their parts. FEBS Lett 2024; 598:2765-2773. [PMID: 38627103 PMCID: PMC11586588 DOI: 10.1002/1873-3468.14869] [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: 10/02/2023] [Revised: 12/15/2023] [Accepted: 01/18/2024] [Indexed: 11/26/2024]
Abstract
Globally, the human population is aging, with an increased proportion of people in "old age" (over 60 years). This trend leads to a growing demand in aging research, stimulating studies in animal models such as mice, fish, and invertebrates. Recently, we published a research summary on the aging of hematopoietic stem cells (HSCs) in C57BL/6 mice based on 12 gene expression datasets. Here, I discuss in greater detail the added value of taking an integrated view, rather than considering each publication separately, to determine genes involved in aging. Considerable variation exists between lists of differentially expressed (DE) genes in HSCs, comparing young and old mice. This variation can result from factors such as inconsistent definitions of "young" and "old", technical variations and variations between laboratory mouse strains. We previously demonstrated that the variation between gene lists could be circumvented by forming a unified list of DE genes-the "aging list"-with citation indexes attached. The most frequently detected DE genes [approximately 200 most cited, which we named the "aging signature" (AS)] were highly consistent across publications. Gene Ontology classification of the AS list identified additional sources of variation between studies: one comes from the specifics of how the data are collected and analyzed; another comes from inconsistencies between how we define the gene categories. As discussed, overcoming these variations is the next challenge toward an integral approach to our systematic knowledge of the aging process.
Collapse
Affiliation(s)
- Leonid V. Bystrykh
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center of Groningen (UMCG)University of GroningenThe Netherlands
| |
Collapse
|
4
|
Chen X, Huang S, Wang L, Liu K, Wu H. Maternal exposure to polystyrene nanoplastics induces sex-specific cardiotoxicity in offspring mice. Heliyon 2024; 10:e39139. [PMID: 39640785 PMCID: PMC11620075 DOI: 10.1016/j.heliyon.2024.e39139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Globally, plastic pollution threatens human health, particularly affecting the hearts of offspring exposed to maternal environmental factors early in development. Few studies have specifically addressed sex-specific cardiac injury in offspring resulting from maternal exposure to polystyrene nanoplastics (PS-NPs). This study investigates the potential cardiac injury in offspring following maternal exposure to 1 mg/L PS-NPs. Pregnant C57BL/6J mice were exposed to PS-NPs until 3 weeks postpartum to establish a maternal exposure model. Heart tissues were collected and weighed, and the transcriptomes of the offspring hearts were sequenced and analyzed using high-throughput RNA sequencing. Immunohistochemical staining was performed to assess the effects of PS-NPs on cardiac immune infiltration, fibrosis, and apoptosis in the offspring. PS-NPs caused a significant reduction in heart and body weight in female offspring compared to males. Additionally, PS-NPs induced sex-specific transcriptional reprogramming and metabolic disruptions in the offspring. PS-NPs also induced significant fibrosis, apoptosis, and increased CD68+ macrophage infiltration in offspring hearts. Notably, PS-NPs induced distinct cardiovascular diseases in the offspring. Fluid shear stress and atherosclerosis were significantly enriched in PS-NP-treated male offspring, while viral myocarditis was predominantly enriched in PS-NP-treated females. Our findings suggest that PS-NPs induce cardiotoxicity in offspring by disrupting metabolism, impairing immunity, and triggering fibrosis and apoptosis, with sex-specific differences. This study provides novel insights and a foundation for understanding sex-specific pharmacological differences and interventions in PS-NP-induced cardiovascular disease in offspring.
Collapse
Affiliation(s)
- Xiuli Chen
- Department of Gynecology and Obstetrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine of Henan University, Zhengzhou, Henan, 450003, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Li Wang
- Department of Gynecology and Obstetrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine of Henan University, Zhengzhou, Henan, 450003, China
| | - Kan Liu
- Department of Gynecology and Obstetrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine of Henan University, Zhengzhou, Henan, 450003, China
| | - Haiying Wu
- Department of Gynecology and Obstetrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine of Henan University, Zhengzhou, Henan, 450003, China
| |
Collapse
|
5
|
Luciano A, Churchill GA. Quantifying the Impact of Co-Housing on Murine Aging Studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606373. [PMID: 39149237 PMCID: PMC11326161 DOI: 10.1101/2024.08.06.606373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Analysis of preclinical lifespan studies often assume that outcome data from co-housed animals are independent. In practice, treatments, such as controlled feeding or putative life-extending compounds, are applied to whole housing units, and as a result the outcomes are potentially correlated within housing units. We consider intra-class (here, intra-cage) correlation in three published and two unpublished lifespan studies of aged mice encompassing more than 20 thousand observations. We show that the independence assumption underlying common analytic techniques does not hold in these data, particularly for traits associated with frailty. We describe and demonstrate various analytical tools available to accommodate this study design and highlight a limitation of standard variance components models (i.e., linear mixed models) which are the usual statistical tool for handling correlated errors. Through simulations, we examine the statistical biases resulting from intra-cage correlations with similar magnitudes as observed in these case studies and discuss implications for power and reproducibility.
Collapse
|
6
|
Yuan R, Adlimoghaddam A, Zhu Y, Han X, Bartke A. Early Life Interventions: Impact on Aging and Longevity. Aging Dis 2024:AD.202.0516. [PMID: 39325935 DOI: 10.14336/ad.202.0516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/05/2024] [Indexed: 09/28/2024] Open
Abstract
Across mammals, lifespans vary remarkably, spanning over a hundredfold difference. Comparative studies consistently reveal a strong inverse relationship between developmental pace and lifespan, hinting at the potential for early-life interventions (ELIs) to influence aging and lifespan trajectories. Focusing on postnatal interventions in mice, this review explores how ELIs influence development, lifespan, and the underlying mechanisms. Previous ELI studies have employed a diverse array of approaches, including dietary modifications, manipulations of the somatotropic axis, and various chemical treatments. Notably, these interventions have demonstrated significant impacts on aging and lifespan in mice. The underlying mechanisms likely involve pathways related to mitochondrial function, mTOR and AMPK signaling, cellular senescence, and epigenetic alterations. Interestingly, ELI studies may serve as valuable models for investigating the complex regulatory mechanisms of development and aging, particularly regarding the interplay among somatic growth, sexual maturation, and lifespan. In addition, prior research has highlighted the intricacies of experimental design and data interpretation. Factors such as timing, sex-specific effects, administration methods, and animal husbandry practices must be carefully considered to ensure the reliability and reproducibility of results, as well as rigorous interpretation. Addressing these factors is essential for advancing our understanding of how development, aging, and lifespan are regulated, potentially opening avenues for interventions that promote healthy aging.
Collapse
Affiliation(s)
- Rong Yuan
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Aida Adlimoghaddam
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Neurology, Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Yun Zhu
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Xiuqi Han
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Andrzej Bartke
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| |
Collapse
|
7
|
Zhu Y, Engmann M, Medina D, Han X, Das P, Bartke A, Ellsworth BS, Yuan R. Metformin treatment of juvenile mice alters aging-related developmental and metabolic phenotypes in sex-dependent and sex-independent manners. GeroScience 2024; 46:3197-3218. [PMID: 38227136 PMCID: PMC11009201 DOI: 10.1007/s11357-024-01067-6] [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: 05/30/2023] [Accepted: 12/30/2023] [Indexed: 01/17/2024] Open
Abstract
Metformin has attracted increasing interest for its potential benefits in extending healthspan and longevity. This study examined the effects of early-life metformin treatment on the development and metabolism of C57BL/6 J (B6) mice, with metformin administered to juvenile mice from 15 to 56 days of age. Metformin treatment led to decreased body weight in both sexes (P < 0.05, t-test). At 9 weeks of age, mice were euthanized and organ weights were recorded. The relative weight of retroperitoneal fat was decreased in females, while relative weights of perigonadal and retroperitoneal fat were decreased, and relative liver weight was increased in males (P < 0.05, t-test). Glucose and insulin tolerance tests (GTT and ITT) were conducted at the age of 7 weeks. ANOVA revealed a significant impairment in insulin sensitivity by the treatment, and a significantly interactive effect on glucose tolerance between sex and treatment, underscoring a disparity in GTT between sexes in response to the treatment. Metformin treatment reduced circulating insulin levels in fasting and non-fasting conditions for male mice, with no significant alterations observed in female mice. qRT-PCR analysis of glucose metabolism-related genes (Akt2, Glut2, Glut4, Irs1, Nrip1, Pi3k, Pi3kca, Pkca) in the liver and skeletal muscle reveals metformin-induced sex- and organ-specific effects on gene expression. Comparison with previous studies in heterogeneous UM-HET3 mice receiving the same treatment suggests that genetic differences may contribute to variability in the effects of metformin treatment on development and metabolism. These findings indicate that early-life metformin treatment affects development and metabolism in both sex- and genetics-dependent manners.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Morgan Engmann
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Xiuqi Han
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Pratyusa Das
- Department of Physiology, Southern Illinois University SIU School of Medicine, 1135 Lincoln Drive, Life Science III, Room 2062, Carbondale, IL, 62901, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA
| | - Buffy S Ellsworth
- Department of Physiology, Southern Illinois University SIU School of Medicine, 1135 Lincoln Drive, Life Science III, Room 2062, Carbondale, IL, 62901, USA
| | - Rong Yuan
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62702, USA.
| |
Collapse
|
8
|
Harrison DE, Strong R, Reifsnyder P, Rosenthal N, Korstanje R, Fernandez E, Flurkey K, Ginsburg BC, Murrell MD, Javors MA, Lopez-Cruzan M, Nelson JF, Willcox BJ, Allsopp R, Watumull DM, Watumull DG, Cortopassi G, Kirkland JL, Tchkonia T, Choi YG, Yousefzadeh MJ, Robbins PD, Mitchell JR, Acar M, Sarnoski EA, Bene MR, Salmon A, Kumar N, Miller RA. Astaxanthin and meclizine extend lifespan in UM-HET3 male mice; fisetin, SG1002 (hydrogen sulfide donor), dimethyl fumarate, mycophenolic acid, and 4-phenylbutyrate do not significantly affect lifespan in either sex at the doses and schedules used. GeroScience 2024; 46:795-816. [PMID: 38041783 PMCID: PMC10828146 DOI: 10.1007/s11357-023-01011-0] [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: 08/28/2023] [Accepted: 11/07/2023] [Indexed: 12/03/2023] Open
Abstract
In genetically heterogeneous (UM-HET3) mice produced by the CByB6F1 × C3D2F1 cross, the Nrf2 activator astaxanthin (Asta) extended the median male lifespan by 12% (p = 0.003, log-rank test), while meclizine (Mec), an mTORC1 inhibitor, extended the male lifespan by 8% (p = 0.03). Asta was fed at 1840 ± 520 (9) ppm and Mec at 544 ± 48 (9) ppm, stated as mean ± SE (n) of independent diet preparations. Both were started at 12 months of age. The 90th percentile lifespan for both treatments was extended in absolute value by 6% in males, but neither was significant by the Wang-Allison test. Five other new agents were also tested as follows: fisetin, SG1002 (hydrogen sulfide donor), dimethyl fumarate, mycophenolic acid, and 4-phenylbutyrate. None of these increased lifespan significantly at the dose and method of administration tested in either sex. Amounts of dimethyl fumarate in the diet averaged 35% of the target dose, which may explain the absence of lifespan effects. Body weight was not significantly affected in males by any of the test agents. Late life weights were lower in females fed Asta and Mec, but lifespan was not significantly affected in these females. The male-specific lifespan benefits from Asta and Mec may provide insights into sex-specific aspects of aging.
Collapse
Affiliation(s)
- David E Harrison
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| | - Randy Strong
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Education, and Clinical Center, Geriatric Research, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Peter Reifsnyder
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Nadia Rosenthal
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Ron Korstanje
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Elizabeth Fernandez
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Education, and Clinical Center, Geriatric Research, San Antonio, TX, USA
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Kevin Flurkey
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Brett C Ginsburg
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Meredith D Murrell
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Martin A Javors
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Marisa Lopez-Cruzan
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - James F Nelson
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Physiology, The University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Bradley J Willcox
- John A. Burns School of Medicine, University of Hawai'I at Mānoa, Honolulu, HI, USA
| | - Richard Allsopp
- John A. Burns School of Medicine, University of Hawai'I at Mānoa, Honolulu, HI, USA
| | | | | | - Gino Cortopassi
- Department of Molecular Biosciences, University of California, Davis, CA, USA
| | | | | | | | | | | | | | - Murat Acar
- Department of Basic Medical Sciences, School of Medicine, Koç University, 34450, Istanbul, Turkey
| | - Ethan A Sarnoski
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Michael R Bene
- Department of Molecular Medicine, The University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Adam Salmon
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Education, and Clinical Center, Geriatric Research, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Molecular Medicine, The University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Navasuja Kumar
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Keller A, Bai H, Budinger S, Eliazer S, Hansen M, Konopka AR, Morales-Nebreda L, Najt CP, Prahlad V, Victorelli S, Vorland CJ, Yuan R, Rhoads TW, Mihaylova MM. The Third Annual Symposium of the Midwest Aging Consortium. J Gerontol A Biol Sci Med Sci 2024; 79:glad239. [PMID: 37804247 PMCID: PMC10799755 DOI: 10.1093/gerona/glad239] [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: 08/11/2023] [Indexed: 10/09/2023] Open
Abstract
The geroscience hypothesis suggests that addressing the fundamental mechanisms driving aging biology will prevent or mitigate the onset of multiple chronic diseases, for which the largest risk factor is advanced age. Research that investigates the root causes of aging is therefore of critical importance given the rising healthcare burden attributable to age-related diseases. The third annual Midwest Aging Consortium symposium was convened as a showcase of such research performed by investigators from institutions across the Midwestern United States. This report summarizes the work presented during a virtual conference across topics in aging biology, including immune function in the lung-particularly timely given the Corona Virus Immune Disease-2019 pandemic-along with the role of metabolism and nutrient-regulated pathways in cellular function with age, the influence of senescence on stem cell function and inflammation, and our evolving understanding of the mechanisms underlying observation of sex dimorphism in aging-related outcomes. The symposium focused on early-stage and emerging investigators, while including keynote presentations from leaders in the biology of aging field, highlighting the diversity and strength of aging research in the Midwest.
Collapse
Affiliation(s)
- Andrea Keller
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, Ohio, USA
- Comprehensive Cancer Center, Wexner Medical Center, Arthur G. James Cancer Hospital, Ohio State University, Columbus, Ohio, USA
| | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Susan Eliazer
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Malene Hansen
- Buck Institute for Research on Aging, Novato, California, USA
| | - Adam R Konopka
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Luisa Morales-Nebreda
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Charles P Najt
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Veena Prahlad
- Department of Biology, Aging Mind and Brain Initiative; Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Stella Victorelli
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Colby J Vorland
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, USA
| | - Rong Yuan
- Geriatric Research Division, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Timothy W Rhoads
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Maria M Mihaylova
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, Ohio, USA
- Comprehensive Cancer Center, Wexner Medical Center, Arthur G. James Cancer Hospital, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
10
|
Kolpakov S, Yashkin A, Ukraintseva S, Yashin A, Akushevich I. Genome-Related Mechanisms Contributing to Differences in Alzheimer's Disease Incidence Between White and Black Older US Adults. J Racial Ethn Health Disparities 2024:10.1007/s40615-024-01907-3. [PMID: 38273182 PMCID: PMC11269527 DOI: 10.1007/s40615-024-01907-3] [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: 09/22/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/27/2024]
Abstract
In this manuscript, we leverage a modified GWAS algorithm adapted for use with multidimensional Cox models and data from the Health and Retirement Study to explore how genetic variation influences the size of the disparity in Alzheimer's disease (AD) incidence between older Black and White US adults. We identified four loci that were associated with higher AD incidence levels in older Black adults: (1) rs11077034 (hazard ratio (HR), 4.98) from the RBFOX1 gene; (2) rs7144494 (HR, 1.68) from the HISLA gene; (3) rs7660552 (HR, 3.07) from the SLC25A4 gene; and (4) rs12599485 (HR, 3.181) from the NIP30 gene. The RBFOX1, HISLA, SLC25A4, and NIP30 genes are known to be associated with AD (RBFOX1, NIP30) directly, and also influence the risk of AD risk-related morbidities such as hypertension (RBFOX1, SLC25A4), depression (SLC25A4), and certain cancers (HISLA, SLC25A4). A likely disparity-generating mechanism may lie in endocytosis and abnormal tissue growing mechanisms, depending on inherited gene mutations and the structure of proxies as well as gene-environment interactions with other risk factors such as lifestyle, education level, and access to adequate medical services.
Collapse
Affiliation(s)
- Stanislav Kolpakov
- Social Science Research Institute, Duke University, Durham, NC, 27710, USA.
| | - Arseniy Yashkin
- Social Science Research Institute, Duke University, Durham, NC, 27710, USA
| | | | - Anatoliy Yashin
- Social Science Research Institute, Duke University, Durham, NC, 27710, USA
| | - Igor Akushevich
- Social Science Research Institute, Duke University, Durham, NC, 27710, USA
| |
Collapse
|
11
|
Yuan R, Hascup E, Hascup K, Bartke A. Relationships among Development, Growth, Body Size, Reproduction, Aging, and Longevity - Trade-Offs and Pace-Of-Life. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1692-1703. [PMID: 38105191 PMCID: PMC10792675 DOI: 10.1134/s0006297923110020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 12/19/2023]
Abstract
Relationships of growth, metabolism, reproduction, and body size to the biological process of aging and longevity have been studied for decades and various unifying "theories of aging" have been proposed to account for the observed associations. In general, fast development, early sexual maturation leading to early reproductive effort, as well as production of many offspring, have been linked to shorter lifespans. The relationship of adult body size to longevity includes a remarkable contrast between the positive correlation in comparisons between different species and the negative correlation seen in comparisons of individuals within the same species. We now propose that longevity and presumably also the rate of aging are related to the "pace-of-life." A slow pace-of-life including slow growth, late sexual maturation, and a small number of offspring, predicts slow aging and long life. The fast pace of life (rapid growth, early sexual maturation, and major reproductive effort) is associated with faster aging and shorter life, presumably due to underlying trade-offs. The proposed relationships between the pace-of-life and longevity apply to both inter- and intra-species comparisons as well as to dietary, genetic, and pharmacological interventions that extend life and to evidence for early life programming of the trajectory of aging. Although available evidence suggests the causality of at least some of these associations, much further work will be needed to verify this interpretation and to identify mechanisms that are responsible.
Collapse
Affiliation(s)
- Rong Yuan
- Southern Illinois University School of Medicine, Department of Internal Medicine, Springfield, IL 19628, USA.
| | - Erin Hascup
- Southern Illinois University School of Medicine, Department of Medical, Microbial, Cellular Immunology and Biology, Springfield, IL 19628, USA.
| | - Kevin Hascup
- Southern Illinois University School of Medicine, Department of Medical, Microbial, Cellular Immunology and Biology, Springfield, IL 19628, USA.
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Andrzej Bartke
- Southern Illinois University School of Medicine, Department of Internal Medicine, Springfield, IL 19628, USA.
| |
Collapse
|
12
|
Keele GR, Zhang JG, Szpyt J, Korstanje R, Gygi SP, Churchill GA, Schweppe DK. Global and tissue-specific aging effects on murine proteomes. Cell Rep 2023; 42:112715. [PMID: 37405913 PMCID: PMC10588767 DOI: 10.1016/j.celrep.2023.112715] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/06/2023] [Accepted: 06/13/2023] [Indexed: 07/07/2023] Open
Abstract
Maintenance of protein homeostasis degrades with age, contributing to aging-related decline and disease. Previous studies have primarily surveyed transcriptional aging changes. To define the effects of age directly at the protein level, we perform discovery-based proteomics in 10 tissues from 20 C57BL/6J mice, representing both sexes at adult and late midlife ages (8 and 18 months). Consistent with previous studies, age-related changes in protein abundance often have no corresponding transcriptional change. Aging results in increases in immune proteins across all tissues, consistent with a global pattern of immune infiltration with age. Our protein-centric data reveal tissue-specific aging changes with functional consequences, including altered endoplasmic reticulum and protein trafficking in the spleen. We further observe changes in the stoichiometry of protein complexes with important roles in protein homeostasis, including the CCT/TriC complex and large ribosomal subunit. These data provide a foundation for understanding how proteins contribute to systemic aging across tissues.
Collapse
Affiliation(s)
| | | | - John Szpyt
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Devin K Schweppe
- Department of Genome Sciences, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
13
|
Knufinke M, MacArthur MR, Ewald CY, Mitchell SJ. Sex differences in pharmacological interventions and their effects on lifespan and healthspan outcomes: a systematic review. FRONTIERS IN AGING 2023; 4:1172789. [PMID: 37305228 PMCID: PMC10249017 DOI: 10.3389/fragi.2023.1172789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023]
Abstract
With an increasing aging population, the burden of age-related diseases magnifies. To alleviate this burden, geroprotection has been an area of intense research focus with the development of pharmacological interventions that target lifespan and/or healthspan. However, there are often sex differences, with compounds mostly tested in male animals. Given the importance of considering both sexes in preclinical research, this neglects potential benefits for the female population, as interventions tested in both sexes often show clear sexual dimorphisms in their biological responses. To further understand the prevalence of sex differences in pharmacological geroprotective intervention studies, we performed a systematic review of the literature according to the PRISMA guidelines. Seventy-two studies met our inclusion criteria and were classified into one of five subclasses: FDA-repurposed drugs, novel small molecules, probiotics, traditional Chinese medicine, and antioxidants, vitamins, or other dietary supplements. Interventions were analyzed for their effects on median and maximal lifespan and healthspan markers, including frailty, muscle function and coordination, cognitive function and learning, metabolism, and cancer. With our systematic review, we found that twenty-two out of sixty-four compounds tested were able to prolong both lifespan and healthspan measures. Focusing on the use of female and male mice, and on comparing their outcomes, we found that 40% of studies only used male mice or did not clarify the sex. Notably, of the 36% of pharmacologic interventions that did use both male and female mice, 73% of these studies showed sex-specific outcomes on healthspan and/or lifespan. These data highlight the importance of studying both sexes in the search for geroprotectors, as the biology of aging is not the same in male and female mice. Systematic Review Registration: [website], identifier [registration number].
Collapse
|
14
|
Bickel MA, Csik B, Gulej R, Ungvari A, Nyul-Toth A, Conley SM. Cell non-autonomous regulation of cerebrovascular aging processes by the somatotropic axis. Front Endocrinol (Lausanne) 2023; 14:1087053. [PMID: 36755922 PMCID: PMC9900125 DOI: 10.3389/fendo.2023.1087053] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Age-related cerebrovascular pathologies, ranging from cerebromicrovascular functional and structural alterations to large vessel atherosclerosis, promote the genesis of vascular cognitive impairment and dementia (VCID) and exacerbate Alzheimer's disease. Recent advances in geroscience, including results from studies on heterochronic parabiosis models, reinforce the hypothesis that cell non-autonomous mechanisms play a key role in regulating cerebrovascular aging processes. Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) exert multifaceted vasoprotective effects and production of both hormones is significantly reduced in aging. This brief overview focuses on the role of age-related GH/IGF-1 deficiency in the development of cerebrovascular pathologies and VCID. It explores the mechanistic links among alterations in the somatotropic axis, specific macrovascular and microvascular pathologies (including capillary rarefaction, microhemorrhages, impaired endothelial regulation of cerebral blood flow, disruption of the blood brain barrier, decreased neurovascular coupling, and atherogenesis) and cognitive impairment. Improved understanding of cell non-autonomous mechanisms of vascular aging is crucial to identify targets for intervention to promote cerebrovascular and brain health in older adults.
Collapse
Affiliation(s)
- Marisa A. Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Anna Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Adam Nyul-Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Department of Public Health, Semmelweis University, Budapest, Hungary
- Institute of Biophysics, Biological Research Centre, Eötvös Lorand Research Network (ELKH), Szeged, Hungary
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
15
|
Ward JM, Vogel P, Sundberg JP. Brain and spinal cord lesions in 28 inbred strains of aging mice. Vet Pathol 2022; 59:1047-1055. [PMID: 36062914 DOI: 10.1177/03009858221120009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Brain and spinal cord histopathology findings in male and female 20-month-old mice in a large-scale aging study of 28 inbred Jackson Laboratory mouse strains from 7 genetic families are described. Brain sections from selected strains at 12 and 24 months of age or older were also reviewed. Common lesions include axonal dystrophy in the gracile and/or cuneate nucleus in the sensory tract of the dorsal medulla and in the spinal cord in all strains. Hirano-like bodies were seen in 24/28 strains, and mineralization was observed in the thalamus of 9/28 strains. Less common lesions were also seen in the cerebellum, cerebral cortex, and other brain areas. No brain or spinal cord tumors were found. Evidence of an impairment of the ubiquitin-proteasome system (UPS) and/or suspected autophagy was manifested as medullary axonal dystrophy with intra-axonal granular eosinophilic bodies and LC3B immunohistochemistry in most strains. RIIIS/J, the most severely affected strain, showed moderate axonal dystrophy at 12 months, which progressed to severe lesions at 20 months. Comparative pathology in various species is discussed.
Collapse
Affiliation(s)
- Jerrold M Ward
- The Jackson Laboratory, Bar Harbor, ME.,Global Vet Pathology, Montgomery Village, MD
| | - Peter Vogel
- St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
16
|
Reagan AM, Onos KD, Heuer SE, Sasner M, Howell GR. Improving mouse models for the study of Alzheimer's disease. Curr Top Dev Biol 2022; 148:79-113. [PMID: 35461569 DOI: 10.1016/bs.ctdb.2021.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease whose risk is influenced by genetic and environmental factors. Although a number of pathological hallmarks have been extensively studied over the last several decades, a complete picture of disease initiation and progression remains unclear. We now understand that numerous cell types and systems are involved in AD pathogenesis, and that this cellular profile may present differently for each individual, making the creation of relevant mouse models challenging. However, with increasingly diverse data made available by genome-wide association studies, we can identify and examine new genes and pathways involved in genetic risk for AD, many of which involve vascular health and inflammation. When developing mouse models, it is critical to assess (1) an aging timeline that represents onset and progression in humans, (2) genetic variants and context, (3) environmental factors present in human populations that result in both neuropathological and functional changes-themes that we address in this chapter.
Collapse
Affiliation(s)
| | | | - Sarah E Heuer
- The Jackson Laboratory, Bar Harbor, ME, United States; Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | | | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, United States; Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States.
| |
Collapse
|
17
|
Fisher JL, Jones EF, Flanary VL, Williams AS, Ramsey EJ, Lasseigne BN. Considerations and challenges for sex-aware drug repurposing. Biol Sex Differ 2022; 13:13. [PMID: 35337371 PMCID: PMC8949654 DOI: 10.1186/s13293-022-00420-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/06/2022] [Indexed: 01/09/2023] Open
Abstract
Sex differences are essential factors in disease etiology and manifestation in many diseases such as cardiovascular disease, cancer, and neurodegeneration [33]. The biological influence of sex differences (including genomic, epigenetic, hormonal, immunological, and metabolic differences between males and females) and the lack of biomedical studies considering sex differences in their study design has led to several policies. For example, the National Institute of Health's (NIH) sex as a biological variable (SABV) and Sex and Gender Equity in Research (SAGER) policies to motivate researchers to consider sex differences [204]. However, drug repurposing, a promising alternative to traditional drug discovery by identifying novel uses for FDA-approved drugs, lacks sex-aware methods that can improve the identification of drugs that have sex-specific responses [7, 11, 14, 33]. Sex-aware drug repurposing methods either select drug candidates that are more efficacious in one sex or deprioritize drug candidates based on if they are predicted to cause a sex-bias adverse event (SBAE), unintended therapeutic effects that are more likely to occur in one sex. Computational drug repurposing methods are encouraging approaches to develop for sex-aware drug repurposing because they can prioritize sex-specific drug candidates or SBAEs at lower cost and time than traditional drug discovery. Sex-aware methods currently exist for clinical, genomic, and transcriptomic information [1, 7, 155]. They have not expanded to other data types, such as DNA variation, which has been beneficial in other drug repurposing methods that do not consider sex [114]. Additionally, some sex-aware methods suffer from poorer performance because a disproportionate number of male and female samples are available to train computational methods [7]. However, there is development potential for several different categories (i.e., data mining, ligand binding predictions, molecular associations, and networks). Low-dimensional representations of molecular association and network approaches are also especially promising candidates for future sex-aware drug repurposing methodologies because they reduce the multiple hypothesis testing burden and capture sex-specific variation better than the other methods [151, 159]. Here we review how sex influences drug response, the current state of drug repurposing including with respect to sex-bias drug response, and how model organism study design choices influence drug repurposing validation.
Collapse
Affiliation(s)
- Jennifer L. Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Emma F. Jones
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Victoria L. Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Avery S. Williams
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Elizabeth J. Ramsey
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| |
Collapse
|
18
|
Goedeke L, Murt KN, Di Francesco A, Camporez JP, Nasiri AR, Wang Y, Zhang X, Cline GW, de Cabo R, Shulman GI. Sex- and strain-specific effects of mitochondrial uncoupling on age-related metabolic diseases in high-fat diet-fed mice. Aging Cell 2022; 21:e13539. [PMID: 35088525 PMCID: PMC8844126 DOI: 10.1111/acel.13539] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Mild uncoupling of oxidative phosphorylation is an intrinsic property of all mitochondria and may have evolved to protect cells against the production of damaging reactive oxygen species. Therefore, compounds that enhance mitochondrial uncoupling are potentially attractive anti‐aging therapies; however, chronic ingestion is associated with a number of unwanted side effects. We have previously developed a controlled‐release mitochondrial protonophore (CRMP) that is functionally liver‐directed and promotes oxidation of hepatic triglycerides by causing a subtle sustained increase in hepatic mitochondrial inefficiency. Here, we sought to leverage the higher therapeutic index of CRMP to test whether mild mitochondrial uncoupling in a liver‐directed fashion could reduce oxidative damage and improve age‐related metabolic disease and lifespan in diet‐induced obese mice. Oral administration of CRMP (20 mg/[kg‐day] × 4 weeks) reduced hepatic lipid content, protein kinase C epsilon activation, and hepatic insulin resistance in aged (74‐week‐old) high‐fat diet (HFD)‐fed C57BL/6J male mice, independently of changes in body weight, whole‐body energy expenditure, food intake, or markers of hepatic mitochondrial biogenesis. CRMP treatment was also associated with a significant reduction in hepatic lipid peroxidation, protein carbonylation, and inflammation. Importantly, long‐term (49 weeks) hepatic mitochondrial uncoupling initiated late in life (94–104 weeks), in conjugation with HFD feeding, protected mice against neoplastic disorders, including hepatocellular carcinoma (HCC), in a strain and sex‐specific manner. Taken together, these studies illustrate the complex variation of aging and provide important proof‐of‐concept data to support further studies investigating the use of liver‐directed mitochondrial uncouplers to promote healthy aging in humans.
Collapse
Affiliation(s)
- Leigh Goedeke
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Kelsey N. Murt
- Translational Gerontology Branch Intramural Research Program National Institute on Aging, NIH Baltimore Maryland USA
| | - Andrea Di Francesco
- Translational Gerontology Branch Intramural Research Program National Institute on Aging, NIH Baltimore Maryland USA
| | - João Paulo Camporez
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
- Department of Physiology Ribeirao Preto School of Medicine University of Sao Paulo São Paulo Brazil
| | - Ali R. Nasiri
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Yongliang Wang
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Xian‐Man Zhang
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Gary W. Cline
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Rafael de Cabo
- Translational Gerontology Branch Intramural Research Program National Institute on Aging, NIH Baltimore Maryland USA
| | - Gerald I. Shulman
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
- Department of Cellular and Molecular Physiology Yale School of Medicine New Haven Connecticut USA
| |
Collapse
|
19
|
Zhu Y, Fang Y, Medina D, Bartke A, Yuan R. Metformin treatment of juvenile mice alters aging-related developmental and metabolic phenotypes. Mech Ageing Dev 2022; 201:111597. [PMID: 34780856 PMCID: PMC8755607 DOI: 10.1016/j.mad.2021.111597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/03/2023]
Abstract
Accumulating evidence suggests that the influence on developmental traits might have long-term effects on aging and health later in life. Metformin is a widely used drug for treating type 2 diabetes and is also used for delaying sexual maturation in girls with precocious puberty. The current report focuses on investigating the effects of metformin on development and metabolic traits. Heterogeneous mice (UM-HET3) were treated with i.p. metformin between the ages of 15 and 56 days. Our results show that body weight and food consumption were increased in both sexes, and sexual maturation was delayed in females. Tail length and circulating insulin-like growth factor 1 (IGF1) levels were significantly increased in both sexes. No significant difference was found in insulin tolerance test, but glucose tolerance was significantly reduced in the males. Circulating adiponectin and insulin levels were altered by metformin treatment in a sex-specific manner. Analysis of quantitative insulin sensitivity check index (QUICKI) suggests that metformin treatment increased insulin sensitivity in female pups, but had opposite effect in male pups. This study revealed that early life metformin treatment alters development and metabolism of mice in both sex-specific and non-specific manners. These effects of metformin may have long-term impacts on aging-related traits.
Collapse
Affiliation(s)
- Yun Zhu
- Deparment of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA,Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - Yimin Fang
- Department of Neurology, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - Rong Yuan
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| |
Collapse
|
20
|
Poudel SB, Dixit M, Yildirim G, Cordoba‐Chacon J, Gahete MD, Yuji I, Kirsch T, Kineman RD, Yakar S. Sexual dimorphic impact of adult-onset somatopause on life span and age-induced osteoarthritis. Aging Cell 2021; 20:e13427. [PMID: 34240807 PMCID: PMC8373322 DOI: 10.1111/acel.13427] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/01/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA), the most prevalent joint disease, is a major cause of disability worldwide. Growth hormone (GH) has been suggested to play significant roles in maintaining articular chondrocyte function and ultimately articular cartilage (AC) homeostasis. In humans, the age-associated decline in GH levels was hypothesized to play a role in the etiology of OA. We studied the impact of adult-onset isolated GH deficiency (AOiGHD) on the life span and skeletal integrity including the AC, in 23- to 30-month-old male and female mice on C57/BL6 genetic background. Reductions in GH during adulthood were associated with extended life span and reductions in body temperature in female mice only. However, end-of-life pathology revealed high levels of lymphomas in both sexes, independent of GH status. Skeletal characterization revealed increases in OA severity in AOiGHD mice, evidenced by AC degradation in both femur and tibia, and significantly increased osteophyte formation in AOiGHD females. AOiGHD males showed significant increases in the thickness of the synovial lining cell layer that was associated with increased markers of inflammation (IL-6, iNOS). Furthermore, male AOiGHD showed significant increases in matrix metalloproteinase-13 (MMP-13), p16, and β-galactosidase immunoreactivity in the AC as compared to controls, indicating increased cell senescence. In conclusion, while the life span of AOiGHD females increased, their health span was compromised by high-grade lymphomas and the development of severe OA. In contrast, AOiGHD males, which did not show extended life span, showed an overall low grade of lymphomas but exhibited significantly decreased health span, evidenced by increased OA severity.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- Department of Molecular PathobiologyDavid B. Kriser Dental CenterNew York University College of DentistryNew YorkNYUSA
| | - Manisha Dixit
- Department of Molecular PathobiologyDavid B. Kriser Dental CenterNew York University College of DentistryNew YorkNYUSA
| | - Gozde Yildirim
- Department of Molecular PathobiologyDavid B. Kriser Dental CenterNew York University College of DentistryNew YorkNYUSA
| | - Jose Cordoba‐Chacon
- Section of Endocrinology, Diabetes, and MetabolismDepartment of MedicineUniversity of Illinois at ChicagoChicagoILUSA
- Research and Development DivisionJesse Brown VA Medical CenterChicagoILUSA
| | - Manuel D. Gahete
- Section of Endocrinology, Diabetes, and MetabolismDepartment of MedicineUniversity of Illinois at ChicagoChicagoILUSA
- Research and Development DivisionJesse Brown VA Medical CenterChicagoILUSA
| | - Ikeno Yuji
- Barshop Institute for Longevity and Aging StudiesUTHSCSASan AntonioTXUSA
| | - Thorsten Kirsch
- Department of Orthopaedic SurgeryNYU Grossman School of MedicineNew YorkNYUSA
- Department of Biomedical EngineeringNYU Tandon School of EngineeringNew YorkNYUSA
| | - Rhonda D. Kineman
- Section of Endocrinology, Diabetes, and MetabolismDepartment of MedicineUniversity of Illinois at ChicagoChicagoILUSA
- Research and Development DivisionJesse Brown VA Medical CenterChicagoILUSA
| | - Shoshana Yakar
- Department of Molecular PathobiologyDavid B. Kriser Dental CenterNew York University College of DentistryNew YorkNYUSA
| |
Collapse
|
21
|
The Jackson Laboratory Nathan Shock Center: impact of genetic diversity on aging. GeroScience 2021; 43:2129-2137. [PMID: 34297313 DOI: 10.1007/s11357-021-00421-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/11/2021] [Indexed: 12/25/2022] Open
Abstract
Healthspan is a complex trait, influenced by many genes and environmental factors that accelerate or delay aging, reduce or increase disease risk, and extend or reduce lifespan. Thus, assessing the role of genetic variation in aging requires an experimental strategy capable of modeling the genetic and biological complexity of human populations. The goal of the The Jackson Laboratory Nathan Shock Center (JAX NSC) is to provide research resources and training for geroscience investigators that seek to understand the role of genetics and genetic diversity on the fundamental process of aging and diseases of human aging using the laboratory mouse as a model system. The JAX NSC has available novel, deeply characterized populations of aged mice, performs state-of-the-art phenotyping of age-relevant traits, provides systems genetics analysis of complex data sets, and provides all of these resources to the geroscience community. The aged animal resources, phenotyping capacity, and genetic expertise available through the JAX NSC benefit the geroscience community by fostering cutting-edge, novel lines of research that otherwise would not be possible. Over the past 15 years, the JAX NSC has transformed aging research across the geroscience community, providing aging mouse resources and tissues to researchers. All JAX NSC data and tools are publicly disseminated on the Mouse Phenome Database and the JAX NSC website, thus ensuring that the resources generated and expertise acquired through the Center are readily available to the aging research community. The JAX NSC will continue to enhance its ability to perform innovative research using a mammalian model to illuminate novel genotype-phenotype relationships and provide a rational basis for designing effective risk assessments and therapeutic interventions to boost longevity and disease-free healthspan.
Collapse
|
22
|
Harrison DE, Strong R, Reifsnyder P, Kumar N, Fernandez E, Flurkey K, Javors MA, Lopez‐Cruzan M, Macchiarini F, Nelson JF, Markewych A, Bitto A, Sindler AL, Cortopassi G, Kavanagh K, Leng L, Bucala R, Rosenthal N, Salmon A, Stearns TM, Bogue M, Miller RA. 17-a-estradiol late in life extends lifespan in aging UM-HET3 male mice; nicotinamide riboside and three other drugs do not affect lifespan in either sex. Aging Cell 2021; 20:e13328. [PMID: 33788371 PMCID: PMC8135004 DOI: 10.1111/acel.13328] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/27/2020] [Accepted: 01/12/2021] [Indexed: 01/23/2023] Open
Abstract
In genetically heterogeneous mice produced by the CByB6F1 x C3D2F1 cross, the "non-feminizing" estrogen, 17-α-estradiol (17aE2), extended median male lifespan by 19% (p < 0.0001, log-rank test) and 11% (p = 0.007) when fed at 14.4 ppm starting at 16 and 20 months, respectively. 90th percentile lifespans were extended 7% (p = 0.004, Wang-Allison test) and 5% (p = 0.17). Body weights were reduced about 20% after starting the 17aE2 diets. Four other interventions were tested in males and females: nicotinamide riboside, candesartan cilexetil, geranylgeranylacetone, and MIF098. Despite some data suggesting that nicotinamide riboside would be effective, neither it nor the other three increased lifespans significantly at the doses tested. The 17aE2 results confirm and extend our original reports, with very similar results when started at 16 months compared with mice started at 10 months of age in a prior study. The consistently large lifespan benefit in males, even when treatment is started late in life, may provide information on sex-specific aspects of aging.
Collapse
Affiliation(s)
| | - Randy Strong
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Geriatric ResearchEducation and Clinical CenterSan AntonioTXUSA
- Research ServiceSouth Texas Veterans Health Care SystemSan AntonioTXUSA
- Department of PharmacologyThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | - Navasuja Kumar
- Department of Pathology and Geriatrics CenterUniversity of MichiganAnn ArborMIUSA
| | - Elizabeth Fernandez
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Geriatric ResearchEducation and Clinical CenterSan AntonioTXUSA
- Department of PharmacologyThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | - Martin A. Javors
- Department of PsychiatryThe University of Texas Health Science CenterSan AntonioTXUSA
| | - Marisa Lopez‐Cruzan
- Department of PsychiatryThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | - James F. Nelson
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Department of PhysiologyThe University of Texas Health Science CenterSan AntonioTXUSA
| | - Adrian Markewych
- Department of PathologyUniversity of Washington Medical CenterSeattleWAUSA
| | - Alessandro Bitto
- Department of PathologyUniversity of Washington Medical CenterSeattleWAUSA
| | - Amy L. Sindler
- Department of Health & Human Physiology and Department of BiochemistryUniversity of IowaIowa CityIAUSA
| | - Gino Cortopassi
- Department of Molecular BiosciencesUniversity of CaliforniaDavisCAUSA
| | - Kylie Kavanagh
- Department of Pathology and Internal MedicineWake Forest School of MedicineWinston‐SalemNCUSA
| | - Lin Leng
- Department of Internal MedicineYale UniversityNew HavenConnecticutUSA
| | - Richard Bucala
- Department of Internal MedicineYale UniversityNew HavenConnecticutUSA
| | | | - Adam Salmon
- Barshop Institute for Longevity and Aging StudiesThe University of Texas Health Science CenterSan AntonioTXUSA
- Geriatric ResearchEducation and Clinical CenterSan AntonioTXUSA
- Research ServiceSouth Texas Veterans Health Care SystemSan AntonioTXUSA
- Department of Molecular MedicineThe University of Texas Health Science CenterSan AntonioTXUSA
| | | | | | - Richard A. Miller
- Department of Pathology and Geriatrics CenterUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
23
|
Bergersen KV, Barnes A, Worth D, David C, Wilson EH. Targeted Transcriptomic Analysis of C57BL/6 and BALB/c Mice During Progressive Chronic Toxoplasma gondii Infection Reveals Changes in Host and Parasite Gene Expression Relating to Neuropathology and Resolution. Front Cell Infect Microbiol 2021; 11:645778. [PMID: 33816350 PMCID: PMC8012756 DOI: 10.3389/fcimb.2021.645778] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
Toxoplasma gondii is a resilient parasite that infects a multitude of warm-blooded hosts and results in a lifelong chronic infection requiring continuous responses by the host. Chronic infection is characterized by a balanced immune response and neuropathology that are driven by changes in gene expression. Previous research pertaining to these processes has been conducted in various mouse models, and much knowledge of infection-induced gene expression changes has been acquired through the use of high throughput sequencing techniques in different mouse strains and post-mortem human studies. However, lack of infection time course data poses a prominent missing link in the understanding of chronic infection, and there is still much that is unknown regarding changes in genes specifically relating to neuropathology and resulting repair mechanisms as infection progresses throughout the different stages of chronicity. In this paper, we present a targeted approach to gene expression analysis during T. gondii infection through the use of NanoString nCounter gene expression assays. Wild type C57BL/6 and BALB/c background mice were infected, and transcriptional changes in the brain were evaluated at 14, 28, and 56 days post infection. Results demonstrate a dramatic shift in both previously demonstrated and novel gene expression relating to neuropathology and resolution in C57BL/6 mice. In addition, comparison between BALB/c and C57BL/6 mice demonstrate initial differences in gene expression that evolve over the course of infection and indicate decreased neuropathology and enhanced repair in BALB/c mice. In conclusion, these studies provide a targeted approach to gene expression analysis in the brain during infection and provide elaboration on previously identified transcriptional changes and also offer insights into further understanding the complexities of chronic T. gondii infection.
Collapse
Affiliation(s)
- Kristina V Bergersen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Ashli Barnes
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Danielle Worth
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Clement David
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States.,NanoString Technologies, Seattle, WA, United States
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
24
|
Liu X, Song Z, Li Y, Yao Y, Fang M, Bai C, An P, Chen H, Chen Z, Tang B, Shen J, Gao X, Zhang M, Chen P, Zhang T, Jia H, Liu X, Hou Y, Yang H, Wang J, Wang F, Xu X, Min J, Nie C, Zeng Y. Integrated genetic analyses revealed novel human longevity loci and reduced risks of multiple diseases in a cohort study of 15,651 Chinese individuals. Aging Cell 2021; 20:e13323. [PMID: 33657282 PMCID: PMC7963337 DOI: 10.1111/acel.13323] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 01/16/2021] [Accepted: 01/23/2021] [Indexed: 12/14/2022] Open
Abstract
There is growing interest in studying the genetic contributions to longevity, but limited relevant genes have been identified. In this study, we performed a genetic association study of longevity in a total of 15,651 Chinese individuals. Novel longevity loci, BMPER (rs17169634; p = 7.91 × 10-15 ) and TMEM43/XPC (rs1043943; p = 3.59 × 10-8 ), were identified in a case-control analysis of 11,045 individuals. BRAF (rs1267601; p = 8.33 × 10-15 ) and BMPER (rs17169634; p = 1.45 × 10-10 ) were significantly associated with life expectancy in 12,664 individuals who had survival status records. Additional sex-stratified analyses identified sex-specific longevity genes. Notably, sex-differential associations were identified in two linkage disequilibrium blocks in the TOMM40/APOE region, indicating potential differences during meiosis between males and females. Moreover, polygenic risk scores and Mendelian randomization analyses revealed that longevity was genetically causally correlated with reduced risks of multiple diseases, such as type 2 diabetes, cardiovascular diseases, and arthritis. Finally, we incorporated genetic markers, disease status, and lifestyles to classify longevity or not-longevity groups and predict life span. Our predictive models showed good performance (AUC = 0.86 for longevity classification and explained 19.8% variance of life span) and presented a greater predictive efficiency in females than in males. Taken together, our findings not only shed light on the genetic contributions to longevity but also elucidate correlations between diseases and longevity.
Collapse
Affiliation(s)
- Xiaomin Liu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
- BGI Education Center University of Chinese Academy of Sciences Shenzhen China
| | - Zijun Song
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Yan Li
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yao Yao
- Center for the Study of Aging and Human Development Medical School of Duke University Durham USA
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
| | - Mingyan Fang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Chen Bai
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
- School of Labor and Human Resources Renmin University Beijing China
| | - Peng An
- Beijing Advanced Innovation Center for Food Nutrition and Human Health China Agricultural University Beijing China
| | - Huashuai Chen
- Business School of Xiangtan University Xiangtan China
| | - Zhihua Chen
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Biyao Tang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Juan Shen
- BGI Genomics BGI‐Shenzhen Shenzhen China
| | - Xiaotong Gao
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | | | - Pengyu Chen
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Tao Zhang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Huijue Jia
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Xiao Liu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yong Hou
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Huanming Yang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Jian Wang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Fudi Wang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health China Agricultural University Beijing China
| | - Xun Xu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
- Guangdong Provincial Key Laboratory of Genome Read and Write Shenzhen China
| | - Junxia Min
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Chao Nie
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yi Zeng
- Center for the Study of Aging and Human Development Medical School of Duke University Durham USA
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
| |
Collapse
|