1
|
McGill CJ, Christensen A, Qian W, Thorwald MA, Lugo JG, Namvari S, White OS, Finch CE, Benayoun BA, Pike CJ. Protection against APOE4-associated phenotypes with the longevity-promoting intervention 17α-estradiol in middle-aged male mice. COMMUNICATIONS MEDICINE 2025; 5:222. [PMID: 40494894 PMCID: PMC12152157 DOI: 10.1038/s43856-025-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/30/2025] [Indexed: 06/18/2025] Open
Abstract
BACKGROUND The apolipoprotein ε4 allele (APOE4) is associated with decreased longevity and increased vulnerability to age-related declines and disorders across multiple systems. Interventions that promote healthspan and lifespan represent a promising strategy to attenuate the development of APOE4-associated aging phenotypes. Here, we studied the ability of the longevity-promoting intervention 17α-estradiol (17αE2) to protect against impairments in APOE4 versus the predominant APOE3 genotype using early middle-aged mice with knock-in of human APOE alleles. METHODS Beginning at age 10 months, male APOE3 or APOE4 mice were treated for 20 weeks with 17αE2 or vehicle then compared body-wide for indices of middle-aged phenotypes. RESULTS Across peripheral and neural measures, APOE4 associates with poorer outcomes. Notably, 17αE2 treatment generally improves outcomes in a genotype-dependent manner, favoring APOE4 mice, including reductions in body weight, plasma leptin, hepatic steatosis, learning and memory, and oxidative damage in the brain. Plasma lipidomics and microglial transcriptomics show reductions in genotype-specific differences with 17αE2 treatment. CONCLUSIONS These findings demonstrate that APOE4 promotes systemic and neural aging phenotypes linked to AD and that 17αE2-mediated healthspan actions show a positive APOE4 bias. Collectively, the findings suggest that longevity-promoting interventions may be useful in mitigating deleterious age-related risks associated with the APOE4 genotype.
Collapse
Affiliation(s)
- Cassandra J McGill
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Wenjie Qian
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Max A Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jose Godoy Lugo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Sara Namvari
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Olivia S White
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, USA.
- Cancer Biology Department, USC Keck School of Medicine, Los Angeles, CA, USA.
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA, USA.
- USC Stem Cell Initiative, Los Angeles, CA, USA.
| | - Christian J Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Ahi EP, Panda B, Primmer CR. The hippo pathway: a molecular bridge between environmental cues and pace of life. BMC Ecol Evol 2025; 25:35. [PMID: 40275190 PMCID: PMC12020181 DOI: 10.1186/s12862-025-02378-8] [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: 12/30/2024] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
The pace of life (POL) is shaped by a complex interplay between genetic and environmental factors, influencing growth, maturation, and lifespan across species. The Hippo signaling pathway, a key regulator of organ size and cellular homeostasis, has emerged as a central integrator of environmental cues that modulate POL traits. In this review, we explore how the Hippo pathway links environmental factors-such as temperature fluctuations and dietary energy availability-to molecular mechanisms governing metabolic balance, hormonal signaling, and reproductive timing. Specifically, we highlight the regulatory interactions between the Hippo pathway and metabolic sensors (AMPK, mTOR, SIRT1 and DLK1-Notch), as well as hormonal signals (IGF-1, kisspeptin, leptin, cortisol, thyroid and sex steroids), which together orchestrate key life-history traits, including growth rates, lifespan and sexual maturation, with a particular emphasis on their role in reproductive timing. Furthermore, we consider its role as a potential coordinator of POL-related molecular processes, such as telomere dynamics and epigenetic mechanisms, within a broader regulatory network. By integrating insights from molecular biology and eco-evolutionary perspectives, we propose future directions to dissect the Hippo pathway's role in POL regulation across taxa. Understanding these interactions will provide new perspectives on how organisms adaptively adjust life-history strategies in response to environmental variability.
Collapse
Affiliation(s)
- Ehsan Pashay Ahi
- Organismal and Evolutionary Biology Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9, 00014, Helsinki, Finland.
| | - Bineet Panda
- Organismal and Evolutionary Biology Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9, 00014, Helsinki, Finland
| | - Craig R Primmer
- Organismal and Evolutionary Biology Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9, 00014, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Yang Y, Mayo A, Levy T, Raz N, Shenhar B, Jarosz DF, Alon U. Compression of morbidity by interventions that steepen the survival curve. Nat Commun 2025; 16:3340. [PMID: 40199852 PMCID: PMC11978790 DOI: 10.1038/s41467-025-57807-5] [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: 10/18/2023] [Accepted: 02/26/2025] [Indexed: 04/10/2025] Open
Abstract
Longevity research aims to extend the healthspan while minimizing the duration of disability and morbidity, known as the sickspan. Most longevity interventions in model organisms extend healthspan, but it is not known whether they compress sickspan relative to the lifespan. Here, we present a theory that predicts which interventions compress relative sickspan, based on the shape of the survival curve. Interventions such as caloric restriction that extend mean lifespan while preserving the shape of the survival curve, are predicted to extend the sickspan proportionally, without compressing it. Conversely, a subset of interventions that extend lifespan and steepen the shape of the survival curve are predicted to compress the relative sickspan. We explain this based on the saturating-removal mathematical model of aging, and present evidence from longitudinal health data in mice, Caenorhabditis elegans and Drosophila melanogaster. We apply this theory to identify potential interventions for compressing the sickspan in mice, and to combinations of longevity interventions. This approach offers potential strategies for compressing morbidity and extending healthspan.
Collapse
Affiliation(s)
- Yifan Yang
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
- Center for Interdisciplinary Studies, Westlake University, Hangzhou, Zhejiang, China.
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Naveh Raz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ben Shenhar
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel F Jarosz
- Department of Chemical & Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
4
|
Ungvari A, Gulej R, Patai R, Papp Z, Toth A, Szabó AÁ, Podesser BK, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Maier AB, Csiszar A, Ungvari Z. Sex-specific mechanisms in vascular aging: exploring cellular and molecular pathways in the pathogenesis of age-related cardiovascular and cerebrovascular diseases. GeroScience 2025; 47:301-337. [PMID: 39754010 PMCID: PMC11872871 DOI: 10.1007/s11357-024-01489-2] [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: 11/04/2024] [Accepted: 12/17/2024] [Indexed: 03/04/2025] Open
Abstract
Aging remains the foremost risk factor for cardiovascular and cerebrovascular diseases, surpassing traditional factors in epidemiological significance. This review elucidates the cellular and molecular mechanisms underlying vascular aging, with an emphasis on sex differences that influence disease progression and clinical outcomes in older adults. We discuss the convergence of aging processes at the macro- and microvascular levels and their contributions to the pathogenesis of vascular diseases. Critical analysis of both preclinical and clinical studies reveals significant sex-specific variations in these mechanisms, which could be pivotal in understanding the disparity in disease morbidity and mortality between sexes. The review highlights key molecular pathways, including oxidative stress, inflammation, and autophagy, and their differential roles in the vascular aging of males and females. We argue that recognizing these sex-specific differences is crucial for developing targeted therapeutic strategies aimed at preventing and managing age-related vascular pathologies. The implications for personalized medicine and potential areas for future research are also explored, emphasizing the need for a nuanced approach to the study and treatment of vascular aging.
Collapse
Affiliation(s)
- Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Attila Á Szabó
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health and Exercise Science, University of Oklahoma, Norman, OK, USA
- Reynolds Section of Geriatrics and Palliative Medicine, Department of Medicine, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andrea B Maier
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
- @AgeSingapore, Healthy Longevity Program, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
5
|
Sathiaseelan R, Isola JVV, Santín-Márquez R, Adekunbi D, Fornalik M, Salmon AB, Stout MB. A pilot study evaluating dosing tolerability of 17α-estradiol in male common marmosets (Callithrix jacchus). GeroScience 2025; 47:1005-1017. [PMID: 39107620 PMCID: PMC11872862 DOI: 10.1007/s11357-024-01311-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: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024] Open
Abstract
17α-estradiol extends healthspan and lifespan in male mice without significant feminization or deleterious effects on reproductive function, making it a candidate for human translation. However, studies in animal models that more accurately replicate human physiology are necessary to establish 17α-estradiol dosing standards for clinical trials. This study evaluated the tolerability of 17α-estradiol treatment in the common marmoset over a short treatment duration. We found that male marmosets tolerated two dosing regimens (0.37-0.47 or 0.62-0.72 mg/kg/day) as evidenced by the absence of gastrointestinal distress, changes in vital signs, or overall health conditions. 17α-estradiol treatment mildly decreased body mass, adiposity, and glycosylated hemoglobin, although these changes were not statistically significant in most instances. However, neither dose of 17α-estradiol elicited feminization in our study, thereby suggesting that optimized dosing regimens may provide health benefits without feminization in primates. Additional studies are needed to determine if longer duration treatments would also be nonfeminizing and elicit significant health benefits, which would aid in developing dosing regimens targeting healthy aging in humans.
Collapse
Affiliation(s)
- Roshini Sathiaseelan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Jose V V Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Roberto Santín-Márquez
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Daniel Adekunbi
- Barshop Institute for Longevity & Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Michal Fornalik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Adam B Salmon
- Barshop Institute for Longevity & Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- South Texas Veterans Affairs Medical Center, San Antonio, TX, USA
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
6
|
Camon C, Prescott M, Neyt C, Decourt C, Stout MB, Campbell RE, Garratt M. Systemic metabolic benefits of 17α-estradiol are not exclusively mediated by ERα in glutamatergic or GABAergic neurons. GeroScience 2024; 46:6127-6140. [PMID: 38776045 PMCID: PMC11493872 DOI: 10.1007/s11357-024-01192-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/02/2024] [Indexed: 10/23/2024] Open
Abstract
17α-Estradiol (17αE2), a less-feminising enantiomer of 17β-estradiol, has been shown to prolong lifespan and improve metabolic health in a sex-specific manner in male, but not in female mice. Recent studies have demonstrated the pivotal role of estrogen receptor α (ERα) in mediating the effects of 17αE2 on metabolic health. However, the specific tissues and/or neuronal signalling pathways that 17αE2 acts through remain to be elucidated. ERα expression in glutamatergic and GABAergic neurons (principal excitatory and inhibitory neurons respectively) in the hypothalamus is essential for estradiol signalling. Therefore, we hypothesised that knocking out ERα from one of these neuronal populations would attenuate the established beneficial metabolic effects of 17αE2 in male mice exposed to a high fat diet. To test this hypothesis we used two established brain specific ERα KO models, targeting either glutamatergic or GABAergic neurons (Vglut2/Vgat-ERαKO). We show that both of these ERα KO models exhibit a strong reduction in ERα expression in the arcuate nucleus of the hypothalamus, a control centre for metabolic regulation. Deletion of ERα from GABAergic neurons significantly diminished the effect of 17αE2 on body weight relative to controls, although these animals still show metabolic benefits with 17αE2 treatment. The response to 17αE2 was unaffected by ERα deletion in glutamatergic neurons. Our results support a benefit of 17αE2 treatment in protection against metabolic dysfunction, but these effects do not depend on exclusive ERα expression in glutamatergic and GABAergic neurons and persist when ERα expression is strongly reduced in the arcuate nucleus of the hypothalamus.
Collapse
Affiliation(s)
- Celine Camon
- Department of Anatomy, University of Otago, Dunedin, New Zealand.
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand.
| | - Mel Prescott
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Christine Neyt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Caroline Decourt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Michael Garratt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
7
|
Camon C, Garratt M, Correa SM. Exploring the effects of estrogen deficiency and aging on organismal homeostasis during menopause. NATURE AGING 2024; 4:1731-1744. [PMID: 39672893 PMCID: PMC11785355 DOI: 10.1038/s43587-024-00767-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
Sex hormone signaling declines during aging, from early midlife through menopause, as a consequence of reduced circulating estrogens and decreased receptiveness to these hormones in target tissues. Estrogens preserve energy homeostasis and promote metabolic health via coordinated and simultaneous effects throughout the brain and body. Age-associated loss of estrogen production during menopause has been implicated in a higher risk for metabolic diseases and increased mortality. However, it remains unclear whether age-associated changes in homeostasis are dependent on reduced estrogen signaling during menopause. Although menopausal hormone therapies containing estrogens can alleviate symptoms, concerns about the risks involved have contributed to a broad decline in the use of these approaches. Non-hormonal therapies have emerged that target tissues or pathways with varying levels of selectivity, reducing risk. We summarize here the broad effects of estrogen loss on homeostasis during menopause, current and emerging therapies and opportunities for understanding homeostatic disruptions associated with menopause.
Collapse
Affiliation(s)
- Celine Camon
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael Garratt
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand.
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Corder KM, Hoffman JM, Sogorovic A, Yang Y, Banerjee A, Sun Y, Stout MB, Austad SN. Negative effects of lifespan extending intervention on resilience in mice. PLoS One 2024; 19:e0312440. [PMID: 39570905 PMCID: PMC11581327 DOI: 10.1371/journal.pone.0312440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/07/2024] [Indexed: 11/24/2024] Open
Abstract
One key goal of basic aging research is the development of reliable assays of both current and future health. These assays could dramatically accelerate progress toward developing health-extending interventions by obviating the need for full lifespan studies, especially if they were informative relatively early in life. One potential approach is the assessment of physiological resilience, defined as the ability to recover from an adverse event. Here, using CB6F1 mice, we evaluated four potential resilience assays, each quantifying recovery from a physiological challenge with clear relevance to humans. The challenges were: (1) anesthesia recovery, (2) restoration of hemoglobin levels after a blood draw, (3) speed of wound healing, and (4) survival after pathogen exposure. We evaluated how each changed with age and with interventions known to extend health in males only (17α-estradiol) or both sexes (calorie restriction). We found that three of the four (recovery from anesthesia, blood draw, and pathogen exposure) showed significant and expected age effects, but wound healing did not. None of the three age-sensitive assays responded to the health-extending interventions in the way we expected, and for some assays, including anesthesia response, interventions actually worsened outcomes. Possible explanations are: (1) our interventions were too brief, (2) the ages we evaluated were too young, (3) our assays did not capture important features of organismal resilience, or (4) organismal resilience is not as clearly related to current or future health as hypothesized. Future studies are needed to determine which of these interpretations is valid and to determine whether other resilience metrics may be more informative about current and future health.
Collapse
Affiliation(s)
- Katelynn M. Corder
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Biological and Environmental Sciences, Samford University, Homewood, AL, United States of America
| | - Jessica M. Hoffman
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Biological Sciences, Augusta University, Augusta, GA, United States of America
| | - Anamarija Sogorovic
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Youfeng Yang
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Anisha Banerjee
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Yi Sun
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Life, Health, and Physical Sciences, Gordon College, Wenham, MA, United States of America
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
| | - Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| |
Collapse
|
9
|
Elmansi AM, Miller RA. Oxidative phosphorylation and fatty acid oxidation in slow-aging mice. Free Radic Biol Med 2024; 224:246-255. [PMID: 39153667 DOI: 10.1016/j.freeradbiomed.2024.08.018] [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: 03/25/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Oxidative metabolism declines with aging in humans leading to multiple metabolic ailments and subsequent inflammation. In mice, there is evidence of age-related suppression of fatty acid oxidation and oxidative phosphorylation in the liver, heart, and muscles. Many interventions that extend healthy lifespan of mice have been developed, including genetic, pharmacological, and dietary interventions. In this article, we review the literature on oxidative metabolism changes in response to those interventions. We also discuss the molecular pathways that mediate those changes, and their potential as targets for future longevity interventions.
Collapse
Affiliation(s)
- Ahmed M Elmansi
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA; University of Michigan Geriatrics Center, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA; University of Michigan Geriatrics Center, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Miller RA, Harrison DE, Cortopassi GA, Dehghan I, Fernandez E, Garratt M, Geisler JG, Ginsburg BC, Han ML, Kaczorowski CC, Kumar N, Leiser SF, Lopez-Cruzan M, Milne G, Mitchell JR, Nelson JF, Reifsnyder PC, Salmon AB, Korstanje R, Rosenthal N, Strong R. Lifespan effects in male UM-HET3 mice treated with sodium thiosulfate, 16-hydroxyestriol, and late-start canagliflozin. GeroScience 2024; 46:4657-4670. [PMID: 38753230 PMCID: PMC11336000 DOI: 10.1007/s11357-024-01176-2] [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: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 06/27/2024] Open
Abstract
Genetically heterogeneous UM-HET3 mice born in 2020 were used to test possible lifespan effects of alpha-ketoglutarate (AKG), 2,4-dinitrophenol (DNP), hydralazine (HYD), nebivolol (NEBI), 16α-hydroxyestriol (OH_Est), and sodium thiosulfate (THIO), and to evaluate the effects of canagliflozin (Cana) when started at 16 months of age. OH_Est produced a 15% increase (p = 0.0001) in median lifespan in males but led to a significant (7%) decline in female lifespan. Cana, started at 16 months, also led to a significant increase (14%, p = 0.004) in males and a significant decline (6%, p = 0.03) in females. Cana given to mice at 6 months led, as in our previous study, to an increase in male lifespan without any change in female lifespan, suggesting that this agent may lead to female-specific late-life harm. We found that blood levels of Cana were approximately 20-fold higher in aged females than in young males, suggesting a possible mechanism for the sex-specific disparities in its effects. NEBI was also found to produce a female-specific decline (4%, p = 0.03) in lifespan. None of the other tested drugs provided a lifespan benefit in either sex. These data bring to 7 the list of ITP-tested drugs that induce at least a 10% lifespan increase in one or both sexes, add a fourth drug with demonstrated mid-life benefits on lifespan, and provide a testable hypothesis that might explain the sexual dimorphism in lifespan effects of the SGLT2 inhibitor Cana.
Collapse
Affiliation(s)
- Richard A Miller
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Geriatrics Center, University of Michigan, Ann Arbor, MI, USA.
| | | | | | - Ishmael Dehghan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth Fernandez
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- GRECC, South Texas Veterans Health Care Network, San Antonio, TX, USA
| | - Michael Garratt
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | | | - Brett C Ginsburg
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Melissa L Han
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Catherine C Kaczorowski
- Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Navasuja Kumar
- Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Scott F Leiser
- Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Marisa Lopez-Cruzan
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ginger Milne
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | | | - James F Nelson
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | - Adam B Salmon
- GRECC, South Texas Veterans Health Care Network, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies and Dept of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | - Randy Strong
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
11
|
Moses E, Atlan T, Sun X, Franěk R, Siddiqui A, Marinov GK, Shifman S, Zucker DM, Oron-Gottesman A, Greenleaf WJ, Cohen E, Ram O, Harel I. The killifish germline regulates longevity and somatic repair in a sex-specific manner. NATURE AGING 2024; 4:791-813. [PMID: 38750187 DOI: 10.1038/s43587-024-00632-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024]
Abstract
Classical evolutionary theories propose tradeoffs among reproduction, damage repair and lifespan. However, the specific role of the germline in shaping vertebrate aging remains largely unknown. In this study, we used the turquoise killifish (Nothobranchius furzeri) to genetically arrest germline development at discrete stages and examine how different modes of infertility impact life history. We first constructed a comprehensive single-cell gonadal atlas, providing cell-type-specific markers for downstream phenotypic analysis. We show here that germline depletion-but not arresting germline differentiation-enhances damage repair in female killifish. Conversely, germline-depleted males instead showed an extension in lifespan and rejuvenated metabolic functions. Through further transcriptomic analysis, we highlight enrichment of pro-longevity pathways and genes in germline-depleted male killifish and demonstrate functional conservation of how these factors may regulate longevity in germline-depleted Caenorhabditis elegans. Our results, therefore, demonstrate that different germline manipulation paradigms can yield pronounced sexually dimorphic phenotypes, implying alternative responses to classical evolutionary tradeoffs.
Collapse
Affiliation(s)
- Eitan Moses
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Tehila Atlan
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Xue Sun
- Department of Biochemistry, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Roman Franěk
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, University of South Bohemia in Ceske Budejovice, Vodnany, Czech Republic
| | - Atif Siddiqui
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University School of Medicine, Jerusalem, Israel
| | | | - Sagiv Shifman
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - David M Zucker
- Department of Statistics and Data Science, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adi Oron-Gottesman
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ehud Cohen
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University School of Medicine, Jerusalem, Israel
| | - Oren Ram
- Department of Biochemistry, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Itamar Harel
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel.
| |
Collapse
|
12
|
Moses E, Atlan T, Sun X, Franek R, Siddiqui A, Marinov GK, Shifman S, Zucker DM, Oron-Gottesman A, Greenleaf WJ, Cohen E, Ram O, Harel I. The killifish germline regulates longevity and somatic repair in a sex-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572041. [PMID: 38187630 PMCID: PMC10769255 DOI: 10.1101/2023.12.18.572041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Classical evolutionary theories propose tradeoffs between reproduction, damage repair, and lifespan. However, the specific role of the germline in shaping vertebrate aging remains largely unknown. Here, we use the turquoise killifish ( N. furzeri ) to genetically arrest germline development at discrete stages, and examine how different modes of infertility impact life-history. We first construct a comprehensive single-cell gonadal atlas, providing cell-type-specific markers for downstream phenotypic analysis. Next, we show that germline depletion - but not arresting germline differentiation - enhances damage repair in female killifish. Conversely, germline-depleted males instead showed an extension in lifespan and rejuvenated metabolic functions. Through further transcriptomic analysis, we highlight enrichment of pro-longevity pathways and genes in germline-depleted male killifish and demonstrate functional conservation of how these factors may regulate longevity in germline-depleted C. elegans . Our results therefore demonstrate that different germline manipulation paradigms can yield pronounced sexually dimorphic phenotypes, implying alternative responses to classical evolutionary tradeoffs.
Collapse
|
13
|
Bubak MP, Mann SN, Borowik AK, Pranay A, Batushansky A, Vieira de Sousa Neto I, Mondal SA, Doidge SM, Davidyan A, Szczygiel MM, Peelor FF, Rigsby S, Broomfield ME, Lacy CI, Rice HC, Stout MB, Miller BF. 17α-Estradiol alleviates high-fat diet-induced inflammatory and metabolic dysfunction in skeletal muscle of male and female mice. Am J Physiol Endocrinol Metab 2024; 326:E226-E244. [PMID: 38197793 PMCID: PMC11193529 DOI: 10.1152/ajpendo.00215.2023] [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: 07/13/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
17α-estradiol (17α-E2) is a naturally occurring nonfeminizing diastereomer of 17β-estradiol that has life span-extending effects in rodent models. To date, studies of the systemic and tissue-specific benefits of 17α-E2 have largely focused on the liver, brain, and white adipose tissue with far less focus on skeletal muscle. Skeletal muscle has an important role in metabolic and age-related disease. Therefore, this study aimed to determine whether 17α-E2 treatment has positive, tissue-specific effects on skeletal muscle during a high-fat feeding. We hypothesized that male, but not female, mice, would benefit from 17α-E2 treatment during a high-fat diet (HFD) with changes in the mitochondrial proteome to support lipid oxidation and subsequent reductions in diacylglycerol (DAG) and ceramide content. To test this hypothesis, we used a multiomics approach to determine changes in lipotoxic lipid intermediates, metabolites, and proteins related to metabolic homeostasis. Unexpectedly, we found that 17α-E2 had marked, but different, beneficial effects within each sex. In male mice, we show that 17α-E2 alleviates HFD-induced metabolic detriments of skeletal muscle by reducing the accumulation of diacylglycerol (DAG), and inflammatory cytokine levels, and altered the abundance of most of the proteins related to lipolysis and β-oxidation. Similar to male mice, 17α-E2 treatment reduced fat mass while protecting muscle mass in female mice but had little muscle inflammatory cytokine levels. Although female mice were resistant to HFD-induced changes in DAGs, 17α-E2 treatment induced the upregulation of six DAG species. In female mice, 17α-E2 treatment changed the relative abundance of proteins involved in lipolysis, β-oxidation, as well as structural and contractile proteins but to a smaller extent than male mice. These data demonstrate the metabolic benefits of 17α-E2 in skeletal muscle of male and female mice and contribute to the growing literature of the use of 17α-E2 for multi tissue health span benefits.NEW & NOTEWORTHY Using a multiomics approach, we show that 17α-E2 alleviates HFD-induced metabolic detriments in skeletal muscle by altering bioactive lipid intermediates, inflammatory cytokines, and the abundance of proteins related to lipolysis and muscle contraction. The positive effects of 17α-E2 in skeletal muscle occur in both sexes but differ in their outcome.
Collapse
Affiliation(s)
- Matthew P Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Shivani N Mann
- Department of Neuroscience, University of Arizona, Tucson, Arizona, United States
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheba, Israel
| | - Ivo Vieira de Sousa Neto
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Samim A Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Stephen M Doidge
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Arik Davidyan
- Department of Biological Sciences, California State University, Sacramento, California, United States
| | - Marcelina M Szczygiel
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Sandra Rigsby
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Matle E Broomfield
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Charles I Lacy
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, United States
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, United States
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma, United States
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
14
|
Isola JVV, Veiga GB, de Brito CRC, Alvarado-Rincón JA, Garcia DN, Zanini BM, Hense JD, Vieira AD, Garratt M, Gasperin BG, Schneider A, Stout MB. 17α-estradiol does not adversely affect sperm parameters or fertility in male mice: implications for reproduction-longevity trade-offs. GeroScience 2023; 45:2109-2120. [PMID: 35689785 PMCID: PMC10651587 DOI: 10.1007/s11357-022-00601-8] [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: 04/21/2022] [Accepted: 06/04/2022] [Indexed: 11/30/2022] Open
Abstract
17α-estradiol (17α-E2) is referred to as a nonfeminizing estrogen that was recently found to extend healthspan and lifespan in male, but not female, mice. Despite an abundance of data indicating that 17α-E2 attenuates several hallmarks of aging in male rodents, very little is known with regard to its effects on feminization and fertility. In these studies, we evaluated the effects of 17α-E2 on several markers of male reproductive health in two independent cohorts of mice. In alignment with our previous reports, chronic 17α-E2 treatment prevented gains in body mass, but did not adversely affect testes mass or seminiferous tubule morphology. We subsequently determined that chronic 17α-E2 treatment also did not alter plasma 17β-estradiol or estrone concentrations, while mildly increasing plasma testosterone levels. We also determined that chronic 17α-E2 treatment did not alter plasma follicle-stimulating hormone or luteinizing hormone concentrations, which suggests 17α-E2 treatment does not alter gonadotropin-releasing hormone neuronal function. Sperm quantity, morphology, membrane integrity, and various motility measures were also unaffected by chronic 17α-E2 treatment in our studies. Lastly, two different approaches were used to evaluate male fertility in these studies. We found that chronic 17α-E2 treatment did not diminish the ability of male mice to impregnate female mice, or to generate successfully implanted embryos in the uterus. We conclude that chronic treatment with 17α-E2 at the dose most commonly employed in aging research does not adversely affect reproductive fitness in male mice, which suggests 17α-E2 does not extend lifespan or curtail disease parameters through tradeoff effects with reproduction.
Collapse
Affiliation(s)
- José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Gabriel B Veiga
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil
| | - Camila R C de Brito
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Joao A Alvarado-Rincón
- Facultad de Ciencias Agropecuarias, Universidad de La Salle, Campus Utopía, Yopal, Casanare, Colombia
| | - Driele N Garcia
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil
| | - Bianka M Zanini
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jéssica D Hense
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil
| | - Arnaldo D Vieira
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Michael Garratt
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Bernardo G Gasperin
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Augusto Schneider
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil.
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
15
|
Mendez Garcia MF, Matsuzaki S, Batushansky A, Newhardt R, Kinter C, Jin Y, Mann SN, Stout MB, Gu H, Chiao YA, Kinter M, Humphries KM. Increased cardiac PFK-2 protects against high-fat diet-induced cardiomyopathy and mediates beneficial systemic metabolic effects. iScience 2023; 26:107131. [PMID: 37534142 PMCID: PMC10391959 DOI: 10.1016/j.isci.2023.107131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/27/2023] [Accepted: 06/10/2023] [Indexed: 08/04/2023] Open
Abstract
A healthy heart adapts to changes in nutrient availability and energy demands. In metabolic diseases like type 2 diabetes (T2D), increased reliance on fatty acids for energy production contributes to mitochondrial dysfunction and cardiomyopathy. A principal regulator of cardiac metabolism is 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2), which is a central driver of glycolysis. We hypothesized that increasing PFK-2 activity could mitigate cardiac dysfunction induced by high-fat diet (HFD). Wild type (WT) and cardiac-specific transgenic mice expressing PFK-2 (GlycoHi) were fed a low fat or HFD for 16 weeks to induce metabolic dysfunction. Metabolic phenotypes were determined by measuring mitochondrial bioenergetics and performing targeted quantitative proteomic and metabolomic analysis. Increasing cardiac PFK-2 had beneficial effects on cardiac and mitochondrial function. Unexpectedly, GlycoHi mice also exhibited sex-dependent systemic protection from HFD, including increased glucose homeostasis. These findings support improving glycolysis via PFK-2 activity can mitigate mitochondrial and functional changes that occur with metabolic syndrome.
Collapse
Affiliation(s)
- Maria F. Mendez Garcia
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Satoshi Matsuzaki
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ryan Newhardt
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Caroline Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Shivani N. Mann
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kenneth M. Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
16
|
Mondal SA, Mann SN, van der Linden C, Sathiaseelan R, Kamal M, Das S, Bubak MP, Logan S, Miller BF, Stout MB. Metabolic benefits of 17α-estradiol in liver are partially mediated by ERβ in male mice. Sci Rep 2023; 13:9841. [PMID: 37330610 PMCID: PMC10276872 DOI: 10.1038/s41598-023-37007-1] [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: 03/25/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Metabolic dysfunction underlies several chronic diseases. Dietary interventions can reverse metabolic declines and slow aging but remaining compliant is difficult. 17α-estradiol (17α-E2) treatment improves metabolic parameters and slows aging in male mice without inducing significant feminization. We recently reported that estrogen receptor α is required for the majority of 17α-E2-mediated benefits in male mice, but that 17α-E2 also attenuates fibrogenesis in liver, which is regulated by estrogen receptor β (ERβ)-expressing hepatic stellate cells (HSC). The current studies sought to determine if 17α-E2-mediated benefits on systemic and hepatic metabolism are ERβ-dependent. We found that 17α-E2 treatment reversed obesity and related systemic metabolic sequela in both male and female mice, but this was partially blocked in female, but not male, ERβKO mice. ERβ ablation in male mice attenuated 17α-E2-mediated benefits on hepatic stearoyl-coenyzme A desaturase 1 (SCD1) and transforming growth factor β1 (TGF-β1) production, which play critical roles in HSC activation and liver fibrosis. We also found that 17α-E2 treatment suppresses SCD1 production in cultured hepatocytes and hepatic stellate cells, indicating that 17α-E2 directly signals in both cell-types to suppress drivers of steatosis and fibrosis. We conclude that ERβ partially controls 17α-E2-mediated benefits on systemic metabolic regulation in female, but not male, mice, and that 17α-E2 likely signals through ERβ in HSCs to attenuate pro-fibrotic mechanisms.
Collapse
Affiliation(s)
- Samim Ali Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Shivani N Mann
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Carl van der Linden
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Roshini Sathiaseelan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Maria Kamal
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Snehasis Das
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Matthew P Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Sreemathi Logan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
17
|
Bubak MP, Mann SN, Borowik AK, Pranay A, Batushansky A, Mondal SA, Diodge SM, Davidyan A, Szczygiel MM, Peelor FR, Rigsby S, Broomfield M, Lacy CI, Rice HC, Stout MB, Miller BF. 17α-estradiol Alleviates High-Fat Diet-Induced Inflammatory and Metabolic Dysfunction in Skeletal Muscle of Male and Female Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542870. [PMID: 37398463 PMCID: PMC10312580 DOI: 10.1101/2023.05.30.542870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Skeletal muscle has a central role in maintaining metabolic homeostasis. 17α-estradiol (17α-E2), a naturally-occurring non-feminizing diastereomer of 17β-estradiol that demonstrates efficacy for improving metabolic outcomes in male, but not female, mice. Despite several lines of evidence showing that 17α-E2 treatment improves metabolic parameters in middle-aged obese and old male mice through effects in brain, liver, and white adipose tissue little is known about how 17α-E2 alters skeletal muscle metabolism, and what role this may play in mitigating metabolic declines. Therefore, this study aimed to determine if 17α-E2 treatment improves metabolic outcomes in skeletal muscle from obese male and female mice following chronic high fat diet (HFD) administration. We hypothesized that male, but not female, mice, would benefit from 17α-E2 treatment during HFD. To test this hypothesis, we used a multi-omics approach to determine changes in lipotoxic lipid intermediates, metabolites, and proteins related to metabolic homeostasis. In male mice, we show that 17α-E2 alleviates HFD-induced metabolic detriments of skeletal muscle by reducing the accumulation of diacylglycerol (DAGs) and ceramides, inflammatory cytokine levels, and reduced the abundance of most of the proteins related to lipolysis and beta-oxidation. In contrast to males, 17α-E2 treatment in female mice had little effect on the DAGs and ceramides content, muscle inflammatory cytokine levels, or changes to the relative abundance of proteins involved in beta-oxidation. These data support to the growing evidence that 17α-E2 treatment could be beneficial for overall metabolic health in male mammals.
Collapse
|
18
|
Isola JVV, Ko S, Ocañas SR, Stout MB. Role of Estrogen Receptor α in Aging and Chronic Disease. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230005. [PMID: 37425648 PMCID: PMC10327608 DOI: 10.20900/agmr20230005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Estrogen receptor alpha (ERα) plays a crucial role in reproductive function in both sexes. It also mediates cellular responses to estrogens in multiple nonreproductive organ systems, many of which regulate systemic metabolic homeostasis and inflammatory processes in mammals. The loss of estrogens and/or ERα agonism during aging is associated with the emergence of several comorbid conditions, particularly in females undergoing the menopausal transition. Emerging data also suggests that male mammals likely benefit from ERα agonism if done in a way that circumvents feminizing characteristics. This has led us, and others, to speculate that tissue-specific ERα agonism may hold therapeutic potential for curtailing aging and chronic disease burden in males and females that are at high-risk of cancer and/or cardiovascular events with traditional estrogen replacement therapies. In this mini-review, we emphasize the role of ERα in the brain and liver, summarizing recent evidence that indicates these two organs systems mediate the beneficial effects of estrogens on metabolism and inflammation during aging. We also discuss how 17α-estradiol administration elicits health benefits in an ERα-dependent manner, which provides proof-of-concept that ERα may be a druggable target for attenuating aging and age-related disease burden.
Collapse
Affiliation(s)
- José V. V. Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
19
|
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: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [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
Affiliation(s)
| | | | - Collin Y. Ewald
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
20
|
Sathiaseelan R, Ahn B, Stout M, Logan S, Wanagat J, Nguyen H, Hord N, Vandiver A, Selvarani R, Ranjit R, Yarbrough H, Masingale A, Miller B, Wolf R, Austad S, Richardson A. A Genetically Heterogeneous Rat Model with Divergent Mitochondrial Genomes. J Gerontol A Biol Sci Med Sci 2023; 78:771-779. [PMID: 36762848 PMCID: PMC10172978 DOI: 10.1093/gerona/glad056] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Indexed: 02/11/2023] Open
Abstract
We generated a genetically heterogenous rat model by a 4-way cross strategy using 4 inbred strains (Brown Norway [BN], Fischer 344 [F344], Lewis [LEW], and Wistar Kyoto [KY]) to provide investigators with a highly genetically diverse rat model from commercially available inbred rats. We made reciprocal crosses between males and females from the 2 F1 hybrids to generate genetically heterogeneous rats with mitochondrial genomes from either the BN (OKC-HETB, a.k.a "B" genotype) or WKY (OKC-HETW a.k.a "W" genotype) parental strains. These two mitochondrial genomes differ at 94 nucleotides, more akin to human mitochondrial genome diversity than that available in classical laboratory mouse strains. Body weights of the B and W genotypes were similar. However, mitochondrial genotype antagonistically affected grip strength and treadmill endurance in females only. In addition, mitochondrial genotype significantly affected multiple responses to a high-fat diet (HFD) and treatment with 17α-estradiol. Contrary to findings in mice in which males only are affected by 17α-estradiol supplementation, female rats fed a HFD beneficially responded to 17α-estradiol treatment as evidenced by declines in body mass, adiposity, and liver mass. Male rats, by contrast, differed in a mitochondrial genotype-specific manner, with only B males responding to 17α-estradiol treatment. Mitochondrial genotype and sex differences were also observed in features of brain-specific antioxidant response to a HFD and 17α-estradiol as shown by hippocampal levels of Sod2 acetylation, JNK, and FoxO3a. These results emphasize the importance of mitochondrial genotype in assessing responses to putative interventions in aging processes.
Collapse
Affiliation(s)
- Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Bumsoo Ahn
- Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma, USA
| | - Sreemathi Logan
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jonathan Wanagat
- Divisions of Geriatrics and Dermatology, Department of Medicine, University of California Los Angeles and Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Hoang Van M Nguyen
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Norman G Hord
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Amy R Vandiver
- Divisions of Geriatrics and Dermatology, Department of Medicine, University of California Los Angeles and Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Ramasamy Selvarani
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Rojina Ranjit
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Hannah Yarbrough
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Anthony Masingale
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Benjamin F Miller
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma, USA
| | - Roman F Wolf
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma, USA
| | - Steven N Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Arlan Richardson
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma, USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
21
|
Mondal SA, Mann SN, van der Linden C, Sathiaseelan R, Kamal M, Das S, Bubak MP, Logan S, Miller BF, Stout MB. Metabolic benefits of 17α-estradiol in liver are partially mediated by ERβ in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.25.534216. [PMID: 36993459 PMCID: PMC10055366 DOI: 10.1101/2023.03.25.534216] [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: 04/13/2023]
Abstract
Metabolic dysfunction underlies several chronic diseases. Dietary interventions can reverse metabolic declines and slow aging but remaining compliant is difficult. 17α-estradiol (17α-E2) treatment improves metabolic parameters and slows aging in male mice without inducing significant feminization. We recently reported that estrogen receptor α is required for the majority of 17α-E2-mediated benefits in male mice, but that 17α-E2 also attenuates fibrogenesis in liver, which is regulated by estrogen receptor β (ERβ)-expressing hepatic stellate cells (HSC). The current studies sought to determine if 17α-E2-mediated benefits on systemic and hepatic metabolism are ERβ-dependent. We found that 17α-E2 treatment reversed obesity and related systemic metabolic sequela in both male and female mice, but this was partially blocked in female, but not male, ERβKO mice. ERβ ablation in male mice attenuated 17α-E2-mediated benefits on hepatic stearoyl-coenyzme A desaturase 1 (SCD1) and transforming growth factor β1 (TGF-β1) production, which play critical roles in HSC activation and liver fibrosis. We also found that 17α-E2 treatment suppresses SCD1 production in cultured hepatocytes and hepatic stellate cells, indicating that 17α-E2 directly signals in both cell-types to suppress drivers of steatosis and fibrosis. We conclude that ERβ partially controls 17α-E2-mediated benefits on systemic metabolic regulation in female, but not male, mice, and that 17α-E2 likely signals through ERβ in HSCs to attenuate pro-fibrotic mechanisms.
Collapse
Affiliation(s)
- Samim Ali Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Shivani N. Mann
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Carl van der Linden
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Roshini Sathiaseelan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Maria Kamal
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Snehasis Das
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Matthew P. Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sreemathi Logan
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Benjamin F. Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
22
|
Ali Mondal S, Sathiaseelan R, Mann SN, Kamal M, Luo W, Saccon TD, Isola JVV, Peelor FF, Li T, Freeman WM, Miller BF, Stout MB. 17α-estradiol, a lifespan-extending compound, attenuates liver fibrosis by modulating collagen turnover rates in male mice. Am J Physiol Endocrinol Metab 2023; 324:E120-E134. [PMID: 36516471 PMCID: PMC9902223 DOI: 10.1152/ajpendo.00256.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Estrogen signaling is protective against chronic liver diseases, although men and a subset of women are contraindicated for chronic treatment with 17β-estradiol (17β-E2) or combination hormone replacement therapies. We sought to determine if 17α-estradiol (17α-E2), a naturally occurring diastereomer of 17β-E2, could attenuate liver fibrosis. We evaluated the effects of 17α-E2 treatment on collagen synthesis and degradation rates using tracer-based labeling approaches in male mice subjected to carbon tetrachloride (CCl4)-induced liver fibrosis. We also assessed the effects of 17α-E2 on markers of hepatic stellate cell (HSC) activation, collagen cross-linking, collagen degradation, and liver macrophage content and polarity. We found that 17α-E2 significantly reduced collagen synthesis rates and increased collagen degradation rates, which was mirrored by declines in transforming growth factor β1 (TGF-β1) and lysyl oxidase-like 2 (LOXL2) protein content in liver. These improvements were associated with increased matrix metalloproteinase 2 (MMP2) activity and suppressed stearoyl-coenzyme A desaturase 1 (SCD1) protein levels, the latter of which has been linked to the resolution of liver fibrosis. We also found that 17α-E2 increased liver fetuin-A protein, a strong inhibitor of TGF-β1 signaling, and reduced proinflammatory macrophage activation and cytokines expression in the liver. We conclude that 17α-E2 reduces fibrotic burden by suppressing HSC activation and enhancing collagen degradation mechanisms. Future studies will be needed to determine if 17α-E2 acts directly in hepatocytes, HSCs, and/or immune cells to elicit these benefits.
Collapse
Affiliation(s)
- Samim Ali Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Roshini Sathiaseelan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Shivani N Mann
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| | - Maria Kamal
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wenyi Luo
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Tatiana D Saccon
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - José V V Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Tiangang Li
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Willard M Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| |
Collapse
|
23
|
Functions of Steroid Hormones in the Male Reproductive Tract as Revealed by Mouse Models. Int J Mol Sci 2023; 24:ijms24032748. [PMID: 36769069 PMCID: PMC9917565 DOI: 10.3390/ijms24032748] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Steroid hormones are capable of diffusing through cell membranes to bind with intracellular receptors to regulate numerous physiological processes. Three classes of steroid hormones, namely androgens, estrogens and glucocorticoids, contribute to the development of the reproductive system and the maintenance of fertility. During the past 30 years, mouse models have been produced in which the expression of genes encoding steroid hormone receptors has been enhanced, partially compromised or eliminated. These mouse models have revealed many of the physiological processes regulated by androgens, estrogens and to a more limited extent glucocorticoids in the testis and male accessory organs. In this review, advances provided by mouse models that have facilitated a better understanding of the molecular regulation of testis and reproductive tract processes by steroid hormones are discussed.
Collapse
|
24
|
Protasoni M, Serrano M. Targeting Mitochondria to Control Ageing and Senescence. Pharmaceutics 2023; 15:352. [PMID: 36839673 PMCID: PMC9960816 DOI: 10.3390/pharmaceutics15020352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/24/2023] Open
Abstract
Ageing is accompanied by a progressive impairment of cellular function and a systemic deterioration of tissues and organs, resulting in increased vulnerability to multiple diseases. Here, we review the interplay between two hallmarks of ageing, namely, mitochondrial dysfunction and cellular senescence. The targeting of specific mitochondrial features in senescent cells has the potential of delaying or even reverting the ageing process. A deeper and more comprehensive understanding of mitochondrial biology in senescent cells is necessary to effectively face this challenge. Here, we discuss the main alterations in mitochondrial functions and structure in both ageing and cellular senescence, highlighting the differences and similarities between the two processes. Moreover, we describe the treatments available to target these pathways and speculate on possible future directions of anti-ageing and anti-senescence therapies targeting mitochondria.
Collapse
Affiliation(s)
- Margherita Protasoni
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Cambridge Institute of Science, Altos Labs, Granta Park, Cambridge CB21 6GP, UK
| |
Collapse
|
25
|
Zhu L, Lv H, Xiao L, Hou Y, Li W, Ge G, Ai C. Diverse effects of α-/β-estradiol on catalytic activities of human UDP-glucuronosyltransferases (UGT). J Steroid Biochem Mol Biol 2023; 225:106196. [PMID: 36181991 DOI: 10.1016/j.jsbmb.2022.106196] [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: 07/28/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 02/01/2023]
Abstract
β-estradiol (β-E2) and α-estradiol (α-E2) act as an endo- and an exon-estrogen in humans, respectively. There is a structural variation in C17-OH configuration of the two estrogens. UDP-glucuronosyltransferases (UGT) are responsible for termination of activities of a variety of endogenous hormones, clinical drugs, and environmental toxicants. The current study was conducted to investigate the effects of the two estrogens towards catalytic activities of UGTs. It was found that β-E2 could decrease activities of UGT1A9, - 2B4 and - 2B7, with Ki values of a few micro-molars. β-E2 could additionally accelerate the activity of UGT2B17 via promoting enzyme-substrate binding and increasing the turn over number. Comparatively, α-E2 displayed much stronger inhibitory potentials towards UGT2B7 and - 2B4, but showed little influence to UGT1A9 and - 2B17. The Ki values for inhibition of UGT2B7 in glucuronidation of different substrates by α-E2 were in a nanomolar range that is only about 1/100-1/50 of β-E2. UGT2B7 structural model was fatherly constructed to explore the mechanism underlying dramatically different inhibition selectivity of the two estrogens. Compared to β-E2, α-E2 formed more hydrophobic and hydrogen-bonded interactions with the residues in the active pocket. It is concluded that the configuration of E2-17-OH determines the inhibitory potentials towards UGTs. The results are useful in better understanding ligand selectivity of UGTs, as well as in further development of α-E2 in health protection.
Collapse
Affiliation(s)
- Liangliang Zhu
- School of Life Science and Research Center of Aquatic Organism Conservation & Water Ecosystem Restoration, Anqing Normal University, Anqing 246133, China
| | - Hui Lv
- School of Life Science and Research Center of Aquatic Organism Conservation & Water Ecosystem Restoration, Anqing Normal University, Anqing 246133, China
| | - Ling Xiao
- School of Resources and Environment and Key Laboratory of Aqueous Environment Protection & Pollution Control of Yangtze River, Anqing Normal University, Anqing 246133, China
| | - Yanyao Hou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China
| | - Wenjuan Li
- School of Life Science and Research Center of Aquatic Organism Conservation & Water Ecosystem Restoration, Anqing Normal University, Anqing 246133, China
| | - Guangbo Ge
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chunzhi Ai
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
26
|
Bosakova T, Tockstein A, Bosakova Z, Komrskova K. Kinetic Study of 17α-Estradiol Mechanism during Rat Sperm Capacitation. Molecules 2022; 27:molecules27134092. [PMID: 35807338 PMCID: PMC9268409 DOI: 10.3390/molecules27134092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/13/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
17α-Estradiol (αE2) is a natural diastereoisomer of 17β-estradiol (E2). It is well known that αE2 can bind to estrogen receptors. However, its biological activity is less than that of E2 and is species and tissue specific. The goal of our study was to propose the mechanism of αE2 hormonal response in rat sperm during their capacitation in vitro and compare it with a previously studied mouse model. Concentration changes in externally added αE2 during capacitation of rat sperm were monitored by the high-performance liquid chromatographic method with tandem mass spectrometric detection (HPLC-MS/MS). The calculated values of relative concentrations Bt were subjected to kinetic analysis. The findings indicated that αE2 in rat sperm did not trigger autocatalytic reaction, in contrast to the mouse sperm, and that the initiation of the hormone penetration through the sperm plasma membrane was substantially faster in rats.
Collapse
Affiliation(s)
- Tereza Bosakova
- Department of Analytical Chemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2, 128 43 Prague, Czech Republic; (T.B.); (A.T.)
- Laboratory of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Antonin Tockstein
- Department of Analytical Chemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2, 128 43 Prague, Czech Republic; (T.B.); (A.T.)
| | - Zuzana Bosakova
- Department of Analytical Chemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2, 128 43 Prague, Czech Republic; (T.B.); (A.T.)
- Correspondence: (Z.B.); (K.K.); Tel.: +420-221951231 (Z.B.); +420-604855871 (K.K.)
| | - Katerina Komrskova
- Laboratory of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
- Department of Zoology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44 Prague, Czech Republic
- Correspondence: (Z.B.); (K.K.); Tel.: +420-221951231 (Z.B.); +420-604855871 (K.K.)
| |
Collapse
|
27
|
Guo Y, Piasecki J, Swiecicka A, Ireland A, Phillips BE, Atherton PJ, Stashuk D, Rutter MK, McPhee JS, Piasecki M. Circulating testosterone and dehydroepiandrosterone are associated with individual motor unit features in untrained and highly active older men. GeroScience 2022; 44:1215-1228. [PMID: 34862585 PMCID: PMC9213614 DOI: 10.1007/s11357-021-00482-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/28/2021] [Indexed: 10/31/2022] Open
Abstract
Long-term exercise training has been considered as an effective strategy to counteract age-related hormonal declines and minimise muscle atrophy. However, human data relating circulating hormone levels with motor nerve function are scant. The aims of the study were to explore associations between circulating sex hormone levels and motor unit (MU) characteristics in older men, including masters athletes competing in endurance and power events. Forty-three older men (mean ± SD age: 69.9 ± 4.6 years) were studied based on competitive status. The serum concentrations of dehydroepiandrosterone (DHEA), total testosterone (T) and estradiol were quantified using liquid chromatography mass spectrometry. Intramuscular electromyographic signals were recorded from vastus lateralis (VL) during 25% of maximum voluntary isometric contractions and processed to extract MU firing rate (FR), and motor unit potential (MUP) features. After adjusting for athletic status, MU FR was positively associated with DHEA levels (p = 0.019). Higher testosterone and estradiol were associated with lower MUP complexity; these relationships remained significant after adjusting for athletic status (p = 0.006 and p = 0.019, respectively). Circulating DHEA was positively associated with MU firing rate in these older men. Higher testosterone levels were associated with reduced MUP complexity, indicating reduced electrophysiological temporal dispersion, which is related to decreased differences in conduction times along axonal branches and/or MU fibres. Although evident in males only, this work highlights the potential of hormone administration as a therapeutic interventional strategy specifically targeting human motor units in older age.
Collapse
Affiliation(s)
- Yuxiao Guo
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jessica Piasecki
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, UK
| | - Agnieszka Swiecicka
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Alex Ireland
- Department of Sport and Exercise Sciences, Musculoskeletal Science and Sports Medicine Research Centre, Manchester Metropolitan University, Manchester, UK
| | - Bethan E. Phillips
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| | - Philip J. Atherton
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| | - Daniel Stashuk
- Department of Systems Design Engineering, University of Waterloo, Waterloo, ON Canada
| | - Martin K. Rutter
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Diabetes, Endocrinology and Metabolism Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Jamie S. McPhee
- Department of Sport and Exercise Sciences, Musculoskeletal Science and Sports Medicine Research Centre, Manchester Metropolitan University, Manchester, UK
| | - Mathew Piasecki
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
28
|
Role of senescence in the chronic health consequences of COVID-19. Transl Res 2022; 241:96-108. [PMID: 34695606 PMCID: PMC8532377 DOI: 10.1016/j.trsl.2021.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023]
Abstract
While the full impact of COVID-19 is not yet clear, early studies have indicated that upwards of 10% of patients experience COVID-19 symptoms longer than 3 weeks, known as Long-Hauler's Syndrome or PACS (postacute sequelae of SARS-CoV-2 infection). There is little known about risk factors or predictors of susceptibility for Long-Hauler's Syndrome, but older adults are at greater risk for severe outcomes and mortality from COVID-19. The pillars of aging (including cellular senescence, telomere dysfunction, impaired proteostasis, mitochondrial dysfunction, deregulated nutrient sensing, genomic instability, progenitor cell exhaustion, altered intercellular communication, and epigenetic alterations) that contribute to age-related dysfunction and chronic diseases (the "Geroscience Hypothesis") may interfere with defenses against viral infection and consequences of these infections. Heightening of the low-grade inflammation that is associated with aging may generate an exaggerated response to an acute COVID-19 infection. Innate immune system dysfunction that leads to decreased senescent cell removal and/or increased senescent cell formation could contribute to accumulation of senescent cells with both aging and viral infections. These processes may contribute to increased risk for long-term COVID-19 sequelae in older or chronically ill patients. Hence, senolytics and other geroscience interventions that may prolong healthspan and alleviate chronic diseases and multimorbidity linked to fundamental aging processes might be an option for delaying, preventing, or alleviating Long-Hauler's Syndrome.
Collapse
Key Words
- ampk, amp-activated protein kinase
- covid-19, coronavirus disease 2019
- covid-fis, a phase 2 placebo-controlled pilot study in covid-19 of fisetin to alleviate dysfunction and excessive inflammatory response in older adults in nursing homes
- cr, caloric restriction
- fga, facility for geroscience analysis
- icu, intensive care unit
- if, intermittent fasting
- ltcf, long-term care facility
- mcc, multiple chronic conditions
- mers-cov, middle east respiratory syndrome coronavirus
- mtor, mammalian target of rapamycin
- nad+, nicotinamide adenine dinucleotide
- nmn, nicotinamide mononucleotide
- nr, nicotinamide riboside
- pacs, postacute sequalae of sars-cov-2 infection
- pamps, pathogen-associated molecular profile factors
- ros, reactive oxygen species
- sars, severe acute respiratory syndrome
- sars-cov-1, severe acute respiratory syndrome coronavirus 1
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
- sasp, senescence-associated secretory phenotype
- snf, skilled nursing facility
- tgn, translational geroscience network
- who, world health organization
Collapse
|
29
|
Wodrich APK, Scott AW, Shukla AK, Harris BT, Giniger E. The Unfolded Protein Responses in Health, Aging, and Neurodegeneration: Recent Advances and Future Considerations. Front Mol Neurosci 2022; 15:831116. [PMID: 35283733 PMCID: PMC8914544 DOI: 10.3389/fnmol.2022.831116] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
Aging and age-related neurodegeneration are both associated with the accumulation of unfolded and abnormally folded proteins, highlighting the importance of protein homeostasis (termed proteostasis) in maintaining organismal health. To this end, two cellular compartments with essential protein folding functions, the endoplasmic reticulum (ER) and the mitochondria, are equipped with unique protein stress responses, known as the ER unfolded protein response (UPR ER ) and the mitochondrial UPR (UPR mt ), respectively. These organellar UPRs play roles in shaping the cellular responses to proteostatic stress that occurs in aging and age-related neurodegeneration. The loss of adaptive UPR ER and UPR mt signaling potency with age contributes to a feed-forward cycle of increasing protein stress and cellular dysfunction. Likewise, UPR ER and UPR mt signaling is often altered in age-related neurodegenerative diseases; however, whether these changes counteract or contribute to the disease pathology appears to be context dependent. Intriguingly, altering organellar UPR signaling in animal models can reduce the pathological consequences of aging and neurodegeneration which has prompted clinical investigations of UPR signaling modulators as therapeutics. Here, we review the physiology of both the UPR ER and the UPR mt , discuss how UPR ER and UPR mt signaling changes in the context of aging and neurodegeneration, and highlight therapeutic strategies targeting the UPR ER and UPR mt that may improve human health.
Collapse
Affiliation(s)
- Andrew P. K. Wodrich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
- College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Andrew W. Scott
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Arvind Kumar Shukla
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Brent T. Harris
- Department of Pathology, Georgetown University, Washington, DC, United States
- Department of Neurology, Georgetown University, Washington, DC, United States
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
Masters MC, Landay AL, Robbins PD, Tchkonia T, Kirkland JL, Kuchel GA, Niedernhofer LJ, Palella FJ. Chronic HIV Infection and Aging: Application of a Geroscience-Guided Approach. J Acquir Immune Defic Syndr 2022; 89:S34-S46. [PMID: 35015744 PMCID: PMC8751288 DOI: 10.1097/qai.0000000000002858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022]
Abstract
ABSTRACT The ability of virally suppressive antiretroviral therapy use to extend the life span of people with HIV (PWH) implies that the age of PWH will also increase. Among PWH, extended survival comes at a cost of earlier onset and increased rates of aging-associated comorbidities and geriatric syndromes, with persistent inflammation and immune dysregulation consequent to chronic HIV infection and to antiretroviral therapy use contributing to an overall decrease in health span. The geroscience hypothesis proposes that the root causes of most aging-related chronic diseases and conditions is the aging process itself. Hence, therapeutically targeting fundamental aging processes could have a greater impact on alleviating or delaying aging-associated comorbidities than addressing each disease individually. Extending the geroscience hypothesis to PWH, we speculate that targeting basic mechanisms of aging will improve overall health with age. Clinical features and pathophysiologic mechanisms of chronic diseases in PWH qualitatively resemble those seen in older adults without HIV. Therefore, drugs that target any of the pillars of aging, including metformin, rapamycin, and nicotinamide adenine dinucleotide precursors, may also slow the rate of onset of age-associated comorbidities and geriatric syndromes in PWH. Drugs that selectively induce apoptosis of senescent cells, termed senolytics, may also improve health span among PWH. Preliminary evidence suggests that senescent cell burden is increased in PWH, implying that senescent cells are an excellent therapeutic target for extending health span. Recently initiated clinical trials evaluating senolytics in age-related diseases offer insights into the design and potential implementation of similar trials for PWH.
Collapse
Affiliation(s)
- Mary C. Masters
- Department of Medicine, Division of Infectious Diseases, Northwestern University, Chicago, IL
| | - Alan L. Landay
- Department of Internal Medicine, Section of Geriatric Medicine Rush University Medical Center, Chicago, IL
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN; and
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; and
| | | | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Frank J. Palella
- Department of Medicine, Division of Infectious Diseases, Northwestern University, Chicago, IL
| |
Collapse
|
31
|
Wink L, Miller RA, Garcia GG. Rapamycin, Acarbose and 17α-estradiol share common mechanisms regulating the MAPK pathways involved in intracellular signaling and inflammation. Immun Ageing 2022; 19:8. [PMID: 35105357 PMCID: PMC8805398 DOI: 10.1186/s12979-022-00264-1] [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: 11/08/2021] [Accepted: 01/19/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Rapamycin (Rapa), acarbose (ACA), and 17α-estradiol (17aE2, males only) have health benefits that increase lifespan of mice. Little is known about how these three agents alter the network of pathways downstream of insulin/IGF1 signals as well as inflammatory/stress responses. RESULTS ACA, Rapa, and 17aE2 (in males, but not in females) oppose age-related increases in the MEK1- ERK1/2-MNK1/2 cascade, and thus reduce phosphorylation of eIF4E, a key component of cap-dependent translation. In parallel, these treatments (in both sexes) reduce age-related increases in the MEK3-p38MAPK-MK2 pathway, to decrease levels of the acute phase response proteins involved in inflammation. CONCLUSION Each of three drugs converges on the regulation of both the ERK1/2 signaling pathway and the p38-MAPK pathway. The changes induced by treatments in ERK1/2 signaling are seen in both sexes, but the 17aE2 effects are male-specific, consistent with the effects on lifespan. However, the inhibition of age-dependent p38MAPK pathways and acute phase responses is triggered in both sexes by all three drugs, suggesting new approaches to prevention or reversal of age-related inflammatory changes in a clinical setting independent of lifespan effects.
Collapse
Affiliation(s)
- Lily Wink
- grid.214458.e0000000086837370Department of Chemistry, University of Michigan College of Literature Science and The Arts, Ann Arbor, USA
| | - Richard A. Miller
- grid.214458.e0000000086837370Department of Pathology, University of Michigan School of Medicine, Ann Arbor, USA ,grid.214458.e0000000086837370University of Michigan Geriatrics Center, Room 3005 BSRB, Box 2200, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200 USA
| | - Gonzalo G. Garcia
- grid.214458.e0000000086837370Department of Pathology, University of Michigan School of Medicine, Ann Arbor, USA
| |
Collapse
|
32
|
Isola JVV, Zanini BM, Hense JD, Alvarado-Rincón JA, Garcia DN, Pereira GC, Vieira AD, Oliveira TL, Collares T, Gasperin BG, Stout MB, Schneider A. Mild calorie restriction, but not 17α-estradiol, extends ovarian reserve and fertility in female mice. Exp Gerontol 2022; 159:111669. [PMID: 35032571 DOI: 10.1016/j.exger.2021.111669] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 12/28/2022]
Abstract
Calorie restriction (CR) (25-40%) is the most commonly studied strategy for curtailing age-related disease and has also been found to extend reproductive lifespan in female mice. However, the effects of mild CR (10%), which is sustainable, on ovarian aging has not yet been addressed. 17α-estradiol (17α-E2) is another intervention shown to positively modulate healthspan and lifespan in mice but its effects on female reproduction remain unclear. We evaluated the effects of mild CR (10%) and 17α-E2 treatment on ovarian reserve and female fertility over a 24-week period, and compared these effects with the more commonly employed 30% CR regimen. Both 10% and 30% CR elicited positive effects on the preservation of ovarian reserve, whereas 17α-E2 did not alter parameters associated with ovarian function. Following refeeding, both 10% and 30% increased fertility as evidenced by greater pregnancy rates. In aligned with the ovarian reserve data, 17α-E2 also failed to improve fertility. Collectively, these data indicate that 10% CR is effective in preserving ovarian function and fertility, while 17α-E2 does not appear to have therapeutic potential for delaying ovarian aging.
Collapse
Affiliation(s)
- José V V Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| | - Bianka M Zanini
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jessica D Hense
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Joao A Alvarado-Rincón
- Facultad de Ciencias Agropecuarias, Universidad de La Salle, Campus Utopía, Yopal, Casanare, Colombia
| | - Driele N Garcia
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Giulia C Pereira
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Arnaldo D Vieira
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Thais L Oliveira
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Tiago Collares
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| | - Bernardo G Gasperin
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
33
|
Pérez-Aldana BE, Martínez-Magaña JJ, Mayén-Lobo YG, Dávila-Ortiz de Montellano DJ, Aviña-Cervantes CL, Ortega-Vázquez A, Genis-Mendoza AD, Sarmiento E, Soto-Reyes E, Juárez-Rojop IE, Tovilla-Zarate CA, González-Castro TB, Nicolini H, López-López M, Monroy-Jaramillo N. Clozapine Long-Term Treatment Might Reduce Epigenetic Age Through Hypomethylation of Longevity Regulatory Pathways Genes. Front Psychiatry 2022; 13:870656. [PMID: 35664466 PMCID: PMC9157596 DOI: 10.3389/fpsyt.2022.870656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Long-term studies have shown significantly lower mortality rates in patients with continuous clozapine (CLZ) treatment than other antipsychotics. We aimed to evaluate epigenetic age and DNA methylome differences between CLZ-treated patients and those without psychopharmacological treatment. The DNA methylome was analyzed using the Infinium MethylationEPIC BeadChip in 31 CLZ-treated patients with psychotic disorders and 56 patients with psychiatric disorders naive to psychopharmacological treatment. Delta age (Δage) was calculated as the difference between predicted epigenetic age and chronological age. CLZ-treated patients were stratified by sex, age, and years of treatment. Differential methylation sites between both groups were determined using linear regression models. The Δage in CLZ-treated patients was on average lower compared with drug-naive patients for the three clocks analyzed; however, after data-stratification, this difference remained only in male patients. Additional differences were observed in Hannum and Horvath clocks when comparing chronological age and years of CLZ treatment. We identified 44,716 differentially methylated sites, of which 87.7% were hypomethylated in CLZ-treated patients, and enriched in the longevity pathway genes. Moreover, by protein-protein interaction, AMPK and insulin signaling pathways were found enriched. CLZ could promote a lower Δage in individuals with long-term treatment and modify the DNA methylome of the longevity-regulating pathways genes.
Collapse
Affiliation(s)
| | - José Jaime Martínez-Magaña
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Yerye Gibrán Mayén-Lobo
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | | | - Carlos Luis Aviña-Cervantes
- Departamento de Psiquiatría, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Alberto Ortega-Vázquez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Alma Delia Genis-Mendoza
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Emmanuel Sarmiento
- Dirección General, Hospital Psiquiátrico Infantil Juan N Navarro, Mexico City, Mexico
| | - Ernesto Soto-Reyes
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Mexico City, Mexico
| | - Isela Esther Juárez-Rojop
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | | | - Thelma Beatriz González-Castro
- División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Méndez, Mexico
| | - Humberto Nicolini
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico.,Grupo de Estudios Médicos y Familiares Carracci, Mexico City, Mexico
| | - Marisol López-López
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Nancy Monroy-Jaramillo
- Departamento de Genética, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| |
Collapse
|
34
|
Einstein-Nathan Shock Center: translating the hallmarks of aging to extend human health span. GeroScience 2021; 43:2167-2182. [PMID: 34463901 DOI: 10.1007/s11357-021-00428-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022] Open
Abstract
The overarching mission of the Einstein-Nathan Shock Center (E-NSC) is to make scientific discoveries in geroscience, leveraging on the expertise in our center in 6 out of the 7 pillars of aging, and to translate their effects towards drug discovery. The relevance of this basic biology of aging discoveries to humans will be confirmed through the unique gero-human resource at E-NSC. This is achieved through services provided by E-NSC, connectivity among its members, attracting worldwide investigators, and providing them with the opportunities to become future leaders. The two central components of the E-NSC are (a) cutting-edge research programs and (b) unique E-NSC research support cores. E-NSC scientists lead NIH-supported cutting-edge research programs that integrate key hallmarks of aging including proteostasis/autophagy, metabolism/inflammaging, genetic/epigenetics, stem cells/regeneration, and translational aging/longevity. Since the inception of the E-NSC, the well-integrated, collaborative, and innovative nature of the multiple supporting state-of-the-art E-NSC research cores form the bedrock of research success at the E-NSC. The three state-of-the-art E-NSC research cores, (i) Proteostasis of Aging Core (PAC), (ii) the Health Span Core (HSC), and (iii) the Human Multi-Omics Core (HMOC), have allowed impressive expansion of translational biological research programs. Expansion was facilitated through the wealth of data coming from genomics/proteomics and metabolomic analysis on human longevity studies, due to access to a variety of biological samples from elderly subjects in clinical trials with aging-targeting drugs, and new drug design services via the PAC to target the hallmarks of aging.
Collapse
|
35
|
Debarba LK, Jayarathne HSM, Miller RA, Garratt M, Sadagurski M. 17-a-estradiol has sex-specific effects on neuroinflammation that are partly reversed by gonadectomy. J Gerontol A Biol Sci Med Sci 2021; 77:66-74. [PMID: 34309657 DOI: 10.1093/gerona/glab216] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
17-α-estradiol (17aE2) treatment from 4-months of age extends lifespan in male mice and can reduce neuroinflammatory responses in the hypothalamus of 12-month-old males. Although 17aE2 improves longevity in males, female mice are unaffected, suggesting a sexually dimorphic pattern of lifespan regulation. We tested whether the sex-specific effects of 17aE2 on neuroinflammatory responses are affected by gonadal removal and whether hypothalamic changes extend to other brain regions in old age. We show that sex-specific effects of 17aE2 on age-associated gliosis are brain region-specific and are partially dependent on gonadectomy. 17aE2 treatment started at 4 months of age protected 25-month-old males from hypothalamic inflammation. Castration before 17aE2 exposure reduced the effect of 17aE2 on hypothalamic astrogliosis in males. By contrast, sex-specific inhibition of microgliosis generated by 17aE2 was not significantly affected by castration. In the hippocampus, gonadectomy influenced the severity of gliosis and the responsiveness to 17aE2 in a region-dependent manner. The male-specific effects of 17aE2 correlate with increases in hypothalamic ERα expression, specifically in gonadally intact males, consistent with the idea that 17aE2 might act through this receptor. Our results indicate that neuroinflammatory responses to 17aE2 are partially controlled by the presence of sex-specific gonads. Loss of gonadal function and age-associated neuroinflammation could, therefore, influence late-life health and disease onset, leading to sexual dimorphism in both aging and in response to drugs that modify the pace of aging.
Collapse
Affiliation(s)
- Lucas K Debarba
- Department of Biological Sciences, IBio (Integrative Biosciences Center), Wayne State University, Detroit, Michigan, MI
| | - Hashan S M Jayarathne
- Department of Biological Sciences, IBio (Integrative Biosciences Center), Wayne State University, Detroit, Michigan, MI
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI
| | - Michael Garratt
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, NZ
| | - Marianna Sadagurski
- Department of Biological Sciences, IBio (Integrative Biosciences Center), Wayne State University, Detroit, Michigan, MI
| |
Collapse
|
36
|
Kinetic Study of 17α-Estradiol Activity in Comparison with 17β-Estradiol and 17α-Ethynylestradiol. Catalysts 2021. [DOI: 10.3390/catal11050634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
17α-estradiol (αE2), an endogenous stereoisomer of the hormone 17β-estradiol (E2), is capable of binding to estrogen receptors (ER). We aimed to mathematically describe, using experimental data, the possible interactions between αE2 and sperm ER during the process of sperm capacitation and to develop a kinetic model. The goal was to compare the suggested kinetic model with previously published results of ER interactions with E2 and 17α-ethynylestradiol (EE2). The HPLC-MS/MS method was developed to monitor the changes of αE2 concentration during capacitation. The calculated relative concentrations Bt were used for kinetic analysis. Rate constants k and molar ratio n were optimized and used for the construction of theoretical B(t) curves. Modifications in αE2–ER interactions were discovered during comparison with models for E2 and EE2. These new interactions displayed autocatalytic formation of an unstable adduct between the hormone and the cytoplasmic receptors. αE2 accumulates between the plasma membrane lipid bilayer with increasing potential, and when the critical level is reached, αE2 penetrates through the inner layer of the plasma membrane into the cytoplasm. It then rapidly reacts with the ER and creates an unstable adduct. The revealed dynamics of αE2–ER action may contribute to understanding tissue rejuvenation and the cancer-related physiology of αE2 signaling.
Collapse
|
37
|
Sidhom S, Schneider A, Fang Y, McFadden S, Darcy J, Sathiaseelan R, Palmer AK, Steyn FJ, Grillari J, Kopchick JJ, Bartke A, Siddiqi S, Masternak MM, Stout MB. 17α-Estradiol Modulates IGF1 and Hepatic Gene Expression in a Sex-Specific Manner. J Gerontol A Biol Sci Med Sci 2021; 76:778-785. [PMID: 32857104 PMCID: PMC8087270 DOI: 10.1093/gerona/glaa215] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
Aging is the greatest risk factor for most chronic diseases. The somatotropic axis is one of the most conserved biological pathways that regulates aging across species. 17α-Estradiol (17α-E2), a diastereomer of 17β-estradiol (17β-E2), was recently found to elicit health benefits, including improved insulin sensitivity and extend longevity exclusively in male mice. Given that 17β-E2 is known to modulate somatotropic signaling in females through actions in the pituitary and liver, we hypothesized that 17α-E2 may be modulating the somatotropic axis in males, thereby contributing to health benefits. Herein, we demonstrate that 17α-E2 increases hepatic insulin-like growth factor 1 (IGF1) production in male mice without inducing any changes in pulsatile growth hormone (GH) secretion. Using growth hormone receptor knockout (GHRKO) mice, we subsequently determined that the induction of hepatic IGF1 by 17α-E2 is dependent upon GH signaling in male mice, and that 17α-E2 elicits no effects on IGF1 production in female mice. We also determined that 17α-E2 failed to feminize the hepatic transcriptional profile in normal (N) male mice, as evidenced by a clear divergence between the sexes, regardless of treatment. Conversely, significant overlap in transcriptional profiles was observed between sexes in GHRKO mice, and this was unaffected by 17α-E2 treatment. Based on these findings, we propose that 17α-E2 acts as a pleiotropic pathway modulator in male mice by uncoupling IGF1 production from insulin sensitivity. In summary, 17α-E2 treatment upregulates IGF1 production in wild-type (and N) male mice in what appears to be a GH-dependent fashion, while no effects in female IGF1 production are observed following 17α-E2 treatment.
Collapse
Affiliation(s)
- Silvana Sidhom
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rio Grande do Sul, Brazil
| | - Yimin Fang
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Samuel McFadden
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Justin Darcy
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Frederik J Steyn
- University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Australia
| | - Johannes Grillari
- Department of Biotechnology, BOKU – University of Natural Resources and Life Sciences, Vienna, Austria
| | - John J Kopchick
- Edison Biotechnology Institute & Heritage College of Osteopathic Medicine, Ohio University, Athens
| | - Andrzej Bartke
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Shadab Siddiqi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center
- Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center
| |
Collapse
|
38
|
Mann SN, Pitel KS, Nelson-Holte MH, Iwaniec UT, Turner RT, Sathiaseelan R, Kirkland JL, Schneider A, Morris KT, Malayannan S, Hawse JR, Stout MB. 17α-Estradiol prevents ovariectomy-mediated obesity and bone loss. Exp Gerontol 2020; 142:111113. [PMID: 33065227 PMCID: PMC8351143 DOI: 10.1016/j.exger.2020.111113] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Menopause is a natural physiological process in older women that is associated with reduced estrogen production and results in increased risk for obesity, diabetes, and osteoporosis. 17α-estradiol (17α-E2) treatment in males, but not females, reverses several metabolic conditions associated with advancing age, highlighting sexually dimorphic actions on age-related pathologies. In this study we sought to determine if 17α-E2 could prevent ovariectomy (OVX)-mediated detriments on adiposity and bone parameters in females. Eight-week-old female C57BL/6J mice were subjected to SHAM or OVX surgery and received dietary 17α-E2 during a six-week intervention period. We observed that 17α-E2 prevented OVX-induced increases in body weight and adiposity. Similarly, uterine weight and luminal cell thickness were decreased by OVX and prevented by 17α-E2 treatment. Interestingly, 17α-E2 prevented OVX-induced declines in tibial metaphysis cancellous bone. And similarly, 17α-E2 improved bone density parameters in both tibia and femur cancellous bone, primarily in OVX mice. In contrast, to the effects on cancellous bone, cortical bone parameters were largely unaffected by OVX or 17α-E2. In the non-weight bearing lumbar vertebrae, OVX reduced trabecular thickness but not spacing, while 17α-E2 increased trabecular thickness and reduced spacing. Despite this, 17α-E2 did improve bone volume/tissue volume in lumbar vertebrae. Overall, we found that 17α-E2 prevented OVX-induced increases in adiposity and changes in bone mass and architecture, with minimal effects in SHAM-operated mice. We also observed that 17α-E2 rescued uterine tissue mass and lining morphology to control levels without inducing hypertrophy, suggesting that 17α-E2 could be considered as an adjunct to traditional hormone replacement therapies.
Collapse
Affiliation(s)
- Shivani N Mann
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Kevin S Pitel
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| | - Molly H Nelson-Holte
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| | - Urszula T Iwaniec
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA.
| | - Russell T Turner
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA.
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | | | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| | - Katherine T Morris
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | | | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|