1
|
Jeon H, Lee S, Kim Y, Kim Y, Shin S, Lee Y, Kim M, Ko E, Lee E, Song BM, Choi H, Hwang N, Han SE, Hwang B, Kim JW, Oh CM, Fang S. Exercise alters transcriptional profiles of senescence and gut barrier integrity in intestinal crypts of aging mice. NPJ AGING 2025; 11:51. [PMID: 40514364 PMCID: PMC12166050 DOI: 10.1038/s41514-025-00242-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 05/29/2025] [Indexed: 06/16/2025]
Abstract
Senescence is the gradual process of aging in tissues and cells, and a primary cause of aging-associated diseases. Among them, intestinal stem cells (ISCs) experience exhaustion during aging, leading to reduced regenerative capacity in the intestinal crypt, which impairs intestinal function and contributes to systemic health issues. Given the critical role ISCs play in maintaining intestinal homeostasis, preventing their senescence is essential for preserving intestinal function. Among the various strategies proposed to slow cellular senescence, regular exercise has emerged as one of the most well-known and widely accepted interventions. Here, we examined how exercise affects the small intestine in an aging mouse model. Using single-cell RNA sequencing, we found that signaling pathways and gene expression related to DNA replication and cell cycle progression were upregulated in ISCs. Additionally, genes promoting ribosome biogenesis showed increased expression in both ISCs and transit amplifying cells. Exercise also recovered Wnt signaling inhibition, potentially influencing ISC differentiation. Furthermore, exercise increased Reg3g expression in Paneth cells and improved gut barrier function, contrasting with findings from a diet-induced obese mouse model. This suggests that regular exercise helps inhibit the aging of ISCs in multiple ways, contributing to the maintenance of intestinal homeostasis.
Collapse
Affiliation(s)
- Hyeonuk Jeon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Siyeon Lee
- Department of Life Science and Biotechnology, Underwood Division, Underwood International College, Yonsei University, Seoul, Republic of Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yeongmin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Soyeon Shin
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yoseob Lee
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minki Kim
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical center, Dallas, TX, USA
| | - Eunbin Ko
- Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Eunsu Lee
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Brian Min Song
- Hallym University College of Medicine, ChunCheon, Gangwon-do, Republic of Korea
| | - Hojeong Choi
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nahee Hwang
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se-Eun Han
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byungjin Hwang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Woo Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Katsarou A, Papadopoulos G, Moustakas II, Papadopetraki A, Moustogiannis A, Legaki AI, Giannousi E, Agrogiannis G, Pantelis P, Veroutis D, Evangelou K, Kotsinas A, Gorgoulis VG, Philippou A, Koutsilieris M, Chatzigeorgiou A. Exercise mitigates liver senescence but does not outmatch dietary restriction in obesity-related MASLD. Metabolism 2025; 170:156325. [PMID: 40505886 DOI: 10.1016/j.metabol.2025.156325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 06/06/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND The present study aims at deciphering the individual or combined benefits of aerobic exercise and dietary restriction on liver senescence, a state characterized by cell cycle arrest and simultaneous resistance to apoptosis, which is considered an established hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS C57BL6 mice were subjected to a normal diet (ND) for 20 weeks or a high-fat diet (HFD) supplemented with 5 % High-fructose Corn Syrup (HFCS) for 12 weeks, followed by eight-week interventions, including dietary restriction (DR), aerobic exercise (EX), a combination of both (DREX) or continuation of a HFD-HFCS diet without intervention. Biomarkers of senescence were analyzed in terms of their liver mRNA expression levels, while GL13 and p21WAF1/CIP1 immunohistochemical stainings were conducted to examine the levels of senescence-associated lipofuscin and p21WAF1/CIP1 respectively, to finally investigate their relationship with the grade of hepatic steatosis and fibrosis observed in the studied mice. RESULTS DR and DREX groups exhibited significantly reduced features of obesity and MASLD-related hepatic steatosis and fibrosis, to a greater extent than the respective amelioration driven by aerobic exercise only in HFDEX animals. A statistically significant increase of mRNA expression was detected for cyclin-dependent kinase p21WAF1/CIP1 in HFD livers as compared to ND, which was also reversed upon DR-inclusive interventions. Immunohistochemical stainings for GL13 and p21WAF1/CIP1, as well as for p16INK4A confirmed the aforementioned alterations of p21WAF1/CIP1 at the tissular level while also revealed a p16INK4A elevation in HFD livers which was reversed only upon DR/DREX. CONCLUSION Liver senescence is responsive both to exercise and dietary restriction, but its amelioration in the context of MASLD is more robust upon DR-inclusive interventions.
Collapse
Affiliation(s)
- Angeliki Katsarou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Grigorios Papadopoulos
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis I Moustakas
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Argyro Papadopetraki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanasios Moustogiannis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Aigli-Ioanna Legaki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eirini Giannousi
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George Agrogiannis
- 1(st) Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Pavlos Pantelis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitris Veroutis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanassios Kotsinas
- Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; Ninewells Hospital and Medical School, University of Dundee, Dundee, UK; Molecular and Clinical Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK; Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|
3
|
Olinger B, Banarjee R, Dey A, Tsitsipatis D, Tanaka T, Ram A, Nyunt T, Daya GN, Peng Z, Shrivastava M, Cui L, Candia J, Simonsick EM, Gorospe M, Walker KA, Ferrucci L, Basisty N. The secretome of senescent monocytes predicts age-related clinical outcomes in humans. NATURE AGING 2025:10.1038/s43587-025-00877-3. [PMID: 40461807 DOI: 10.1038/s43587-025-00877-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 04/17/2025] [Indexed: 06/11/2025]
Abstract
Cellular senescence increases with age and contributes to age-related declines and pathologies. We identified circulating biomarkers of senescence and related them to clinical traits in humans to facilitate future noninvasive assessment of individual senescence burden, and efficacy testing of novel senotherapeutics. Using a nanoparticle-based proteomic workflow, we profiled the senescence-associated secretory phenotype (SASP) in THP-1 monocytes and examined these proteins in 1,060 plasma samples from the Baltimore Longitudinal Study of Aging. Machine-learning models trained on THP-1 monocyte SASP associated SASP signatures with several age-related phenotypes in a test cohort, including body fat composition, blood lipids, inflammatory markers and mobility-related traits, among others. Notably, a subset of SASP-based predictions, including a high-impact SASP panel, were validated in InCHIANTI, an independent aging cohort. These results demonstrate the clinical relevance of the circulating SASP and identify potential senescence biomarkers that could inform future clinical studies.
Collapse
Affiliation(s)
- Bradley Olinger
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Reema Banarjee
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Amit Dey
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Anjana Ram
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Thedoe Nyunt
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Gulzar N Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Mansi Shrivastava
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Linna Cui
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Julian Candia
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Eleanor M Simonsick
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Nathan Basisty
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA.
| |
Collapse
|
4
|
Wang G, Li G, Song A, Zhao Y, Yu J, Wang Y, Dai W, Salas M, Qin H, Medrano L, Dow J, Li A, Armstrong B, Fueger PT, Yu H, Zhu Y, Shao M, Wu X, Jiang L, Campisi J, Yang X, Wang QA. Distinct adipose progenitor cells emerging with age drive active adipogenesis. Science 2025; 388:eadj0430. [PMID: 40273250 DOI: 10.1126/science.adj0430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 05/30/2024] [Accepted: 02/05/2025] [Indexed: 04/26/2025]
Abstract
Starting at middle age, adults often suffer from visceral adiposity and associated adverse metabolic disorders. Lineage tracing in mice revealed that adipose progenitor cells (APCs) in visceral fat undergo extensive adipogenesis during middle age. Thus, despite the low turnover rate of adipocytes in young adults, adipogenesis is unlocked during middle age. Transplantations quantitatively showed that APCs in middle-aged mice exhibited high adipogenic capacity cell-autonomously. Single-cell RNA sequencing identified a distinct APC population, the committed preadipocyte, age-enriched (CP-A), emerging at this age. CP-As demonstrated elevated proliferation and adipogenesis activity. Pharmacological and genetic manipulations indicated that leukemia inhibitory factor receptor signaling was indispensable for CP-A adipogenesis and visceral fat expansion. These findings uncover a fundamental mechanism of age-dependent adipose remodeling, offering critical insights into age-related metabolic diseases.
Collapse
Affiliation(s)
- Guan Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Gaoyan Li
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anying Song
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yutian Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiayu Yu
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yifan Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Wenting Dai
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Martha Salas
- Light Microscopy Core, City of Hope Medical Center, Duarte, CA, USA
| | - Hanjun Qin
- The Integrative Genomics Core, City of Hope Medical Center, Duarte, CA, USA
| | - Leonard Medrano
- Division of Developmental and Translational Diabetes and Endocrinology Research, City of Hope Medical Center, Duarte, CA, USA
| | - Joan Dow
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, City of Hope Medical Center, Duarte, CA, USA
| | - Aimin Li
- Pathology Core of Shared Resources, City of Hope Medical Center, Duarte, CA, USA
| | - Brian Armstrong
- Light Microscopy Core, City of Hope Medical Center, Duarte, CA, USA
| | - Patrick T Fueger
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Hua Yu
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yi Zhu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Mengle Shao
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Xiwei Wu
- The Integrative Genomics Core, City of Hope Medical Center, Duarte, CA, USA
| | - Lei Jiang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | | | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Qiong A Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| |
Collapse
|
5
|
Zhu F, Lin BR, Lin SH, Yu CH, Yang YM. Hepatic-specific vitamin D receptor downregulation alleviates aging-related metabolic dysfunction-associated steatotic liver disease. World J Gastroenterol 2025; 31:104117. [PMID: 40248374 PMCID: PMC12001193 DOI: 10.3748/wjg.v31.i14.104117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/21/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is defined by the abnormal lipid deposition in hepatocytes. The prevalence of MASLD is significantly increased in the elderly population, suggesting that aging may be related to the occurrence of MASLD. Emerging evidences suggest that vitamin D receptor (VDR) may be implicated in the progression of MASLD. Therefore, additional researches are warranted to elucidate whether VDR plays a role in aging-related MASLD. AIM To investigate the relationship between aging and MASLD and explore the role and related mechanisms of VDR in aging-related MASLD. METHODS Cellular senescence models were established, and the senescence phenotype of telomerase RNA component knockout mice was validated. These mice were then used as a senescence model for subsequent studies. Changes in VDR expression in the livers of aging mice were examined. VDR knockdown models, including cell knockdown models and hepatic-specific VDR knockout mice, were constructed, and MASLD was established in these models. Additionally, vitamin D (VD)-supplemented models, including senescent liver cell lines and senescent mice, were constructed. RESULTS The steatosis in senescent liver cells was more severe than in normal cells (P < 0.05). Moreover, hepatic steatosis was significantly more pronounced in senescence model mice compared to control group when the MASLD model was successfully induced (P < 0.05). Therefore, we concluded that aging aggravated hepatic steatosis. The hepatic expression of VDR increased after aging. VDR knockdown in senescent liver cells and senescent mice alleviated hepatic steatosis (P < 0.05). When senescent liver cells were stimulated with VD, cellular steatosis was aggravated (P < 0.05). However, VD supplementation had no effect on aging mice. CONCLUSION Aging can lead to increased hepatic steatosis, and the hepatic-specific knockdown of VDR alleviated aging-related MASLD. VDR could serve as a potential molecular target for aging-related MASLD.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Bing-Ru Lin
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Shi-Hua Lin
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Chao-Hui Yu
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Yun-Mei Yang
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
- Key Laboratory of Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
6
|
Conover CA, Oxvig C. The IGF System and Aging. Endocr Rev 2025; 46:214-223. [PMID: 39418083 PMCID: PMC11894535 DOI: 10.1210/endrev/bnae029] [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: 07/03/2024] [Revised: 09/20/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
There is strong evidence that IGF signaling is involved in fundamental aspects of the aging process. However, the extracellular part of the IGF system is complex with various receptors, ligand effectors, high-affinity IGF-binding proteins, proteinases, and endogenous inhibitors that all, along with their biological context, must be considered. The IGF system components are evolutionarily conserved, underscoring the importance of understanding this system in physiology and pathophysiology. This review will briefly describe the different components of the IGF system and then discuss past and current literature regarding IGF and aging, with a focus on cellular senescence, model organisms of aging, centenarian genetics, and 3 age-related diseases-pulmonary fibrosis, Alzheimer disease, and macular degeneration-in appropriate murine models and in humans. Commonalities in mechanism suggest conditions where IGF system components may be disease drivers and potential targets in promoting healthy aging in humans.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
7
|
Palmer AK, St. Sauver J, Fielding RA, Atkinson E, White TA, McGree M, Weston S, LeBrasseur NK. The Influence of Body Mass Index on Biomarkers of Cellular Senescence in Older Adults. J Gerontol A Biol Sci Med Sci 2025; 80:glae251. [PMID: 39447036 PMCID: PMC11949428 DOI: 10.1093/gerona/glae251] [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: 03/10/2024] [Indexed: 10/26/2024] Open
Abstract
Obesity accelerates the onset and progression of age-related conditions. In preclinical models, obesity drives cellular senescence, a cell fate that compromises tissue health and function, in part through a robust and diverse secretome. In humans, components of the secretome have been used as senescence biomarkers that are predictive of age-related disease, disability, and mortality. Here, using biospecimens and clinical data from two large and independent cohorts of older adults, we tested the hypothesis that the circulating concentrations of senescence biomarkers are influenced by body mass index. After adjusting for age, sex, and race, we observed significant increases in activin A, Fas, MDC, PAI1, PARC, TNFR1, and VEGFA, and a significant decrease in RAGE, from normal weight, to overweight, to obesity body mass index categories by linear regression in both cohorts (all p < .05). These results highlight the influence of body mass index on circulating concentrations of senescence biomarkers.
Collapse
Affiliation(s)
- Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, College of Medicine, Rochester, Minnesota, USA
- Division of Hospital Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jennifer St. Sauver
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Roger A Fielding
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Elizabeth Atkinson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas A White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, College of Medicine, Rochester, Minnesota, USA
| | - Michaela McGree
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Susan Weston
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, College of Medicine, Rochester, Minnesota, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Conover CA. Cellular senescence and PAPP-A. Growth Horm IGF Res 2025; 80:101637. [PMID: 39904113 DOI: 10.1016/j.ghir.2025.101637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Cellular senescence and its accompanying secretome have major impact on growth and aging of mammalian organisms. A novel protease, PAPP-A, regulates the bioavailability of an important growth factor, insulin-like growth factor (IGF)-I, and has major impact on growth and aging. This short review will explore a new perspective of cellular senescence and PAPP-A.
Collapse
Affiliation(s)
- Cheryl A Conover
- Department of Endocrinology, Endocrine Research Unit, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States of America.
| |
Collapse
|
9
|
Joung J, Heo Y, Kim Y, Kim J, Choi H, Jeon T, Jang Y, Kim EJ, Lee SH, Suh JM, Elledge SJ, Kim MS, Kang C. Cell enlargement modulated by GATA4 and YAP instructs the senescence-associated secretory phenotype. Nat Commun 2025; 16:1696. [PMID: 39962062 PMCID: PMC11833096 DOI: 10.1038/s41467-025-56929-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Dynamic changes in cell size are associated with development and pathological conditions, including aging. Although cell enlargement is a prominent morphological feature of cellular senescence, its functional implications are unknown; moreover, how senescent cells maintain their enlargement state is less understood. Here we show that an extensive remodeling of actin cytoskeleton is necessary for establishing senescence-associated cell enlargement and pro-inflammatory senescence-associated secretory phenotype (SASP). This remodeling is attributed to a balancing act between the SASP regulator GATA4 and the mechanosensor YAP on the expression of the Rho family of GTPase RHOU. Genetic or pharmacological interventions that reduce cell enlargement attenuate SASP with minimal effect on senescence growth arrest. Mechanistically, actin cytoskeleton remodeling couples cell enlargement to the nuclear localization of GATA4 and NF-κB via the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex. RhoU protein accumulates in mouse adipose tissue under senescence-inducing conditions. Furthermore, RHOU expression correlates with SASP expression in adipose tissue during human aging. Thus, our study highlights an unexpected instructive role of cell enlargement in modulating the SASP and reveals a mechanical branch in the senescence regulatory network.
Collapse
Affiliation(s)
- Joae Joung
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Yekang Heo
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Yeonju Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Jaejin Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Haebeen Choi
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Taerang Jeon
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Yeji Jang
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Eun-Jung Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea
| | - Sang Heon Lee
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, South Korea
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, South Korea
| | - Stephen J Elledge
- Department of Genetics, Harvard Medical School and Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Mi-Sung Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea.
| | - Chanhee Kang
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.
- Center for Systems Geroscience, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
10
|
Moaddel R, Candia J, Ubaida-Mohien C, Tanaka T, Moore AZ, Zhu M, Fantoni G, Church S, D'Agostino J, Fan J, Shehadeh N, De S, Lehrmann E, Kaileh M, Simonsick E, Sen R, Egan JM, Ferrucci L. Healthy Aging Metabolomic and Proteomic Signatures Across Multiple Physiological Compartments. Aging Cell 2025:e70014. [PMID: 39952253 DOI: 10.1111/acel.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025] Open
Abstract
The study of biomarkers in biofluids and tissues expanded our understanding of the biological processes that drive physiological and functional manifestations of aging. However, most of these studies were limited to examining one biological compartment, an approach that fails to recognize that aging pervasively affects the whole body. The simultaneous modeling of hundreds of metabolites and proteins across multiple compartments may provide a more detailed picture of healthy aging and point to differences between chronological and biological aging. Herein, we report proteomic analyses of plasma and urine collected in healthy men and women, age 22-92 years. Using these data, we developed a series of metabolomic and proteomic predictors of chronological age for plasma, urine, and skeletal muscle. We then defined a biological aging score, which measures the departure between an individual's predicted age and the expected predicted age for that individual based on the full cohort. We show that these predictors are significantly and independently related to clinical phenotypes important for aging, such as inflammation, iron deficiency anemia, muscle mass, and renal and hepatic functions. Despite a different set of selected biomarkers in each compartment, the different scores reflect a similar degree of deviation from healthy aging in single individuals, thus allowing identification of subjects with significant accelerated or decelerated biological aging.
Collapse
Affiliation(s)
- R Moaddel
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - J Candia
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - C Ubaida-Mohien
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - T Tanaka
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - A Z Moore
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - M Zhu
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - G Fantoni
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - S Church
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - J D'Agostino
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - J Fan
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - N Shehadeh
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - S De
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - E Lehrmann
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - M Kaileh
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - E Simonsick
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - R Sen
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - J M Egan
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - L Ferrucci
- Biomedical Research Centre, National Institute on Aging, NIH, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Al‐Naggar IM, Antony M, Baker D, Wang L, Godoy LDC, Kuo C, Fraser MO, Smith PP, Xu M, Kuchel GA. Polyploid superficial uroepithelial bladder barrier cells express features of cellular senescence across the lifespan and are insensitive to senolytics. Aging Cell 2025; 24:e14399. [PMID: 39644167 PMCID: PMC11822673 DOI: 10.1111/acel.14399] [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: 11/17/2023] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 12/09/2024] Open
Abstract
Lower urinary tract dysfunction (LUTD) increases with aging. Ensuing symptoms including incontinence greatly impact quality of life, isolation, depression, and nursing home admission. The aging bladder is hypothesized to be central to this decline, however, it remains difficult to pinpoint a singular strong driver of aging-related bladder dysfunction. Many molecular and cellular changes occur with aging, contributing to decreased resilience to internal and external stressors, affecting urinary control and exacerbating LUTD. In this study, we examined whether cellular senescence, a cell fate involved in the etiology of most aging diseases, contributes to LUTD. We found that umbrella cells (UCs), luminal barrier uroepithelial cells in the bladder, show senescence features over the mouse lifespan. These polyploid UCs exhibit high cyclin D1 staining, previously reported to mediate tetraploidy-induced senescence in vitro. These senescent UCs were not eliminated by the senolytic combination of Dasatinib and Quercetin. We also tested the effect of a high-fat diet (HFD) and senescent cell transplantation on bladder function and showed that both models induce cystometric changes similar to natural aging in mice, with no effect of senolytics on HFD-induced changes. These findings illustrate the heterogeneity of cellular senescence in varied tissues, while also providing potential insights into the origin of urothelial cancer. We conclude that senescence of bladder uroepithelial cells plays a role in normal physiology, namely in their role as barrier cells, helping promote uroepithelial integrity and impermeability and maintaining the urine-blood barrier.
Collapse
Affiliation(s)
- Iman M. Al‐Naggar
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- Department of Cell BiologyUniversity of Connecticut HealthFarmingtonConnecticutUSA
- Department of SurgeryUniversity of Connecticut HealthFarmingtonConnecticutUSA
| | - Maria Antony
- The University of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Dylan Baker
- Department of Genetics & Genome SciencesUniversity of Connecticut HealthFarmingtonConnecticutUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Lichao Wang
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
| | - Lucas Da Cunha Godoy
- The Cato T. Laurencin Institute for Regenerative EngineeringFarmingtonConnecticutUSA
| | - Chia‐Ling Kuo
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- The Cato T. Laurencin Institute for Regenerative EngineeringFarmingtonConnecticutUSA
| | - Matthew O. Fraser
- Department of Research & DevelopmentDurham Veterans Affairs Medical CentersDurhamNorth CarolinaUSA
| | - Phillip P. Smith
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- Department of SurgeryUniversity of Connecticut HealthFarmingtonConnecticutUSA
- Connecticut Institute for Brain and Cognitive ScienceUniversity of ConnecticutStorrsConnecticutUSA
| | - Ming Xu
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- Department of Genetics & Genome SciencesUniversity of Connecticut HealthFarmingtonConnecticutUSA
| | - George A. Kuchel
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
| |
Collapse
|
12
|
Lyons CE, Pallais JP, McGonigle S, Mansk RP, Collinge CW, Yousefzadeh MJ, Baker DJ, Schrank PR, Williams JW, Niedernhofer LJ, van Deursen JM, Razzoli M, Bartolomucci A. Chronic social stress induces p16-mediated senescent cell accumulation in mice. NATURE AGING 2025; 5:48-64. [PMID: 39528642 DOI: 10.1038/s43587-024-00743-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Life stress can shorten lifespan and increase risk for aging-related diseases, but the biology underlying this phenomenon remains unclear. Here we assessed the effect of chronic stress on cellular senescence-a hallmark of aging. Exposure to restraint stress, a psychological non-social stress model, increased p21Cip1 exclusively in the brains of male, but not female mice, and in a p16Ink4a-independent manner. Conversely, exposure to chronic subordination stress (only males were tested) increased key senescent cell markers in peripheral blood mononuclear cells, adipose tissue and brain, in a p16Ink4a-dependent manner. p16Ink4a-positive cells in the brain of chronic subordination stress-exposed mice were primarily hippocampal and cortical neurons with evidence of DNA damage that could be reduced by p16Ink4a cell clearance. Clearance of p16Ink4a-positive cells was not sufficient to ameliorate the adverse effects of social stress on measured metrics of healthspan. Overall, our findings indicate that social stress induces an organ-specific and p16Ink4a-dependent accumulation of senescent cells, illuminating a fundamental way by which the social environment can contribute to aging.
Collapse
Affiliation(s)
- Carey E Lyons
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Jean Pierre Pallais
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Seth McGonigle
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Rachel P Mansk
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Charles W Collinge
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Paul F. Glenn Center for the Biology of Aging, Mayo Clinic, Rochester, MN, USA
| | - Patricia R Schrank
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jesse W Williams
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
13
|
Zhang Y, Jiang Y, Yang X, Huang Y, Pan A, Liao Y. Adipose tissue senescence: Biological changes, hallmarks and therapeutic approaches. Mech Ageing Dev 2024; 222:111988. [PMID: 39265709 DOI: 10.1016/j.mad.2024.111988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
Adipose tissue (AT), the largest energy storage reservoir and endocrine organ, plays a crucial role in regulating systemic energy metabolism. As one of the most vulnerable tissues during aging, the plasticity of AT is impaired. With age, AT undergoes redistribution, characterized by expansion of visceral adipose tissue (VAT) and reduction of peripheral subcutaneous adipose tissue (SAT). Additionally, age-related changes in AT include reduced adipogenesis of white adipocytes, decreased proliferation and differentiation capacity of mesenchymal stromal/stem cells (MSCs), diminished thermogenic capacity in brown/beige adipocytes, and dysregulation of immune cells. Specific and sensitive hallmarks enable the monitoring and evaluation of the biological changes associated with aging. In this study, we have innovatively proposed seven characteristic hallmarks of AT senescence, including telomere attrition, epigenetic alterations, genomic instability, mitochondrial dysfunction, disabled macroautophagy, cellular senescence, and chronic inflammation, which are intricately interconnected and mutually regulated. Finally, we discussed anti-aging strategies targeting AT, offering insights into mitigating or delaying metabolic disturbances caused by AT senescence.
Collapse
Affiliation(s)
- Yajuan Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Yaoyao Jiang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Xiaoyue Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Yumei Huang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China.
| |
Collapse
|
14
|
Calubag MF, Robbins PD, Lamming DW. A nutrigeroscience approach: Dietary macronutrients and cellular senescence. Cell Metab 2024; 36:1914-1944. [PMID: 39178854 PMCID: PMC11386599 DOI: 10.1016/j.cmet.2024.07.025] [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: 03/22/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Cellular senescence, a process in which a cell exits the cell cycle in response to stressors, is one of the hallmarks of aging. Senescence and the senescence-associated secretory phenotype (SASP)-a heterogeneous set of secreted factors that disrupt tissue homeostasis and promote the accumulation of senescent cells-reprogram metabolism and can lead to metabolic dysfunction. Dietary interventions have long been studied as methods to combat age-associated metabolic dysfunction, promote health, and increase lifespan. A growing body of literature suggests that senescence is responsive to diet, both to calories and specific dietary macronutrients, and that the metabolic benefits of dietary interventions may arise in part through reducing senescence. Here, we review what is currently known about dietary macronutrients' effect on senescence and the SASP, the nutrient-responsive molecular mechanisms that may mediate these effects, and the potential for these findings to inform the development of a nutrigeroscience approach to healthy aging.
Collapse
Affiliation(s)
- Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paul D Robbins
- Institute On the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
15
|
Al-Naggar IM, Campellone KG, Espinoza SE, Justice JN, Orr ME, Kozikowski C, van der Willik O, Thatcher C, Schmader K, Pignolo RJ, Newman JC, Kuchel GA. Bringing Geroscience into the Mainstream: From Education to Clinical Practice, What Will It Take? J Gerontol A Biol Sci Med Sci 2024; 79:glae020. [PMID: 39126344 PMCID: PMC11316222 DOI: 10.1093/gerona/glae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 08/12/2024] Open
Affiliation(s)
- Iman M Al-Naggar
- Center on Aging, UConn Health, Farmington, Connecticut, USA
- Departments of Cell Biology and Surgery, UConn Health, Farmington, Connecticut, USA
| | - Kenneth G Campellone
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Sara E Espinoza
- Department of Medicine, Center for Geroscience, Diabetes and Aging Center, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Jamie N Justice
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- XPRIZE Foundation, Culver City, California, USA
| | - Miranda E Orr
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Chester Kozikowski
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Kenneth Schmader
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Robert J Pignolo
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - John C Newman
- Department of Medicine, University of California in San Francisco, San Francisco, California, USA
- Buck Institute for Research on Aging, Novato, California, USA
| | | |
Collapse
|
16
|
Olinger B, Banarjee R, Dey A, Tsitsipatis D, Tanaka T, Ram A, Nyunt T, Daya G, Peng Z, Cui L, Candia J, Simonsick EM, Gorospe M, Walker KA, Ferrucci L, Basisty N. A plasma proteomic signature links secretome of senescent monocytes to aging- and obesity-related clinical outcomes in humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.01.24311368. [PMID: 39371126 PMCID: PMC11451660 DOI: 10.1101/2024.08.01.24311368] [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: 10/08/2024]
Abstract
Cellular senescence increases with age and contributes to age-related declines and pathologies. We identified circulating biomarkers of senescence associated with diverse clinical traits in humans to facilitate future non-invasive assessment of individual senescence burden and efficacy testing of novel senotherapeutics. Using a novel nanoparticle-based proteomic workflow, we profiled the senescence-associated secretory phenotype (SASP) in monocytes and examined these proteins in plasma samples (N = 1060) from the Baltimore Longitudinal Study of Aging (BLSA). Machine learning models trained on monocyte SASP associated with several age-related phenotypes in a test cohort, including body fat composition, blood lipids, inflammation, and mobility-related traits, among others. Notably, a subset of SASP-based predictions, including a 'high impact' SASP panel that predicts age- and obesity-related clinical traits, were validated in InCHIANTI, an independent aging cohort. These results demonstrate the clinical relevance of the circulating SASP and identify relevant biomarkers of senescence that could inform future clinical studies.
Collapse
Affiliation(s)
- Bradley Olinger
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Reema Banarjee
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Amit Dey
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Anjana Ram
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Thedoe Nyunt
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Gulzar Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Linna Cui
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Eleanor M. Simonsick
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Nathan Basisty
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Tanaka H, Sugawara S, Tanaka Y, Loo TM, Tachibana R, Abe A, Kamiya M, Urano Y, Takahashi A. Dipeptidylpeptidase-4-targeted activatable fluorescent probes visualize senescent cells. Cancer Sci 2024; 115:2762-2773. [PMID: 38802068 PMCID: PMC11309953 DOI: 10.1111/cas.16229] [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: 03/26/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Senescent cells promote cancer development and progression through chronic inflammation caused by a senescence-associated secretory phenotype (SASP). Although various senotherapeutic strategies targeting senescent cells have been developed for the prevention and treatment of cancers, technology for the in vivo detection and evaluation of senescent cell accumulation has not yet been established. Here, we identified activatable fluorescent probes targeting dipeptidylpeptidase-4 (DPP4) as an effective probe for detecting senescent cells through an enzymatic activity-based screening of fluorescent probes. We also determined that these probes were highly, selectively, and rapidly activated in senescent cells during live cell imaging. Furthermore, we successfully visualized senescent cells in the organs of mice using DPP4-targeted probes. These results are expected to lead to the development of a diagnostic technology for noninvasively detecting senescent cells in vivo and could play a role in the application of DPP4 prodrugs for senotherapy.
Collapse
Affiliation(s)
- Hisamichi Tanaka
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
- Department of JFCR Cancer Biology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Sho Sugawara
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Yoko Tanaka
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Tze Mun Loo
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Ryo Tachibana
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Atsuki Abe
- Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mako Kamiya
- Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
- Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Akiko Takahashi
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
- Cancer Cell Communication Project, NEXT‐Ganken ProgramJapanese Foundation for Cancer ResearchTokyoJapan
| |
Collapse
|
18
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
19
|
Xie L, Cheng Y, Hu B, Chen X, An Y, Xia Z, Cai G, Li C, Peng H. PCLAF induces bone marrow adipocyte senescence and contributes to skeletal aging. Bone Res 2024; 12:38. [PMID: 38961077 PMCID: PMC11222446 DOI: 10.1038/s41413-024-00337-5] [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: 10/14/2023] [Revised: 03/30/2024] [Accepted: 04/11/2024] [Indexed: 07/05/2024] Open
Abstract
Bone marrow adipocytes (BMAds) affect bone homeostasis, but the mechanism remains unclear. Here, we showed that exercise inhibited PCNA clamp-associated factor (PCLAF) secretion from the bone marrow macrophages to inhibit BMAds senescence and thus alleviated skeletal aging. The genetic deletion of PCLAF in macrophages inhibited BMAds senescence and delayed skeletal aging. In contrast, the transplantation of PCLAF-mediated senescent BMAds into the bone marrow of healthy mice suppressed bone turnover. Mechanistically, PCLAF bound to the ADGRL2 receptor to inhibit AKT/mTOR signaling that triggered BMAds senescence and subsequently spread senescence among osteogenic and osteoclastic cells. Of note, we developed a PCLAF-neutralizing antibody and showed its therapeutic effects on skeletal health in old mice. Together, these findings identify PCLAF as an inducer of BMAds senescence and provide a promising way to treat age-related osteoporosis.
Collapse
Affiliation(s)
- Lingqi Xie
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yalun Cheng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Xin Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yuze An
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Zhuying Xia
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Guangping Cai
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan, 410008, China
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
20
|
Sanfeliu-Redondo D, Gibert-Ramos A, Gracia-Sancho J. Cell senescence in liver diseases: pathological mechanism and theranostic opportunity. Nat Rev Gastroenterol Hepatol 2024; 21:477-492. [PMID: 38485755 DOI: 10.1038/s41575-024-00913-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 06/30/2024]
Abstract
The liver is not oblivious to the passage of time, as ageing is a major risk factor for the development of acute and chronic liver diseases. Ageing produces alterations in all hepatic cells, affecting their phenotype and function and worsening the prognosis of liver disease. The ageing process also implies the accumulation of a cellular state characterized by a persistent proliferation arrest and a specific secretory phenotype named cellular senescence. Indeed, senescent cells have key roles in many physiological processes; however, their accumulation owing to ageing or pathological conditions contributes to the damage occurring in chronic diseases. The aim of this Review is to provide an updated description of the pathophysiological events in which hepatic senescent cells are involved and their role in liver disease progression. Finally, we discuss novel geroscience therapies that could be applied to prevent or improve liver diseases and age-mediated hepatic deregulations.
Collapse
Affiliation(s)
- David Sanfeliu-Redondo
- Liver Vascular Biology Laboratory, IDIBAPS Biomedical Research Institute - Hospital Clínic de Barcelona & CIBEREHD, Barcelona, Spain
| | - Albert Gibert-Ramos
- Liver Vascular Biology Laboratory, IDIBAPS Biomedical Research Institute - Hospital Clínic de Barcelona & CIBEREHD, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Laboratory, IDIBAPS Biomedical Research Institute - Hospital Clínic de Barcelona & CIBEREHD, Barcelona, Spain.
- Department of Visceral Surgery and Medicine, Inselspital - University of Bern, Bern, Switzerland.
| |
Collapse
|
21
|
Christian LM, Kiecolt-Glaser JK, Cole SW, Burd CE, Madison AA, Wilson SJ, Rosko AE. Psychoneuroimmunology in multiple myeloma and autologous hematopoietic stem cell transplant: Opportunities for research among patients and caregivers. Brain Behav Immun 2024; 119:507-519. [PMID: 38643954 DOI: 10.1016/j.bbi.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024] Open
Abstract
Multiple myeloma (MM) is an incurable cancer and is the leading indication for autologous hematopoietic stem cell transplantation (HSCT). To be eligible for HSCT, a patient must have a caregiver, as caregivers play a central role in HSCT preparation and recovery. MM patients remain on treatment indefinitely, and thus patients and their caregivers face long-term challenges including the intensity of HSCT and perpetual therapy after transplant. Importantly, both patients and their caregivers show heightened depressive and anxiety symptoms, with dyadic correspondence evidenced and caregivers' distress often exceeding that of patients. An extensive psychoneuroimmunology (PNI) literature links distress with health via immune and neuroendocrine dysregulation as well as biological aging. However, data on PNI in the context of multiple myeloma - in patients or caregivers - are remarkably limited. Distress in MM patients has been associated with poorer outcomes including higher inflammation, greater one year post-HSCT hospital readmissions, and worse overall survival. Further, anxiety and depression are linked to biological aging and may contribute to the poor long-term health of both patients and caregivers. Because MM generally affects older adults, individual differences in biological aging may represent an important modifier of MM biology and HSCT treatment outcomes. There are a number of clinical scenarios in which biologically younger people could be prescribed more intensive therapies, with potential for greater benefit, by using a personalized cancer therapy approach based on the quantification of physiologic reserve. Further, despite considerable psychological demands, the effects of distress on health among MM caregivers is largely unexamined. Within this context, the current critical review highlights gaps in knowledge at the intersection of HSCT, inflammation, and biological aging in the context of MM. Research in this area hold promise for opportunities for novel and impactful psychoneuroimmunology (PNI) research to enhance health outcomes, quality of life, and longevity among both MM patients and their caregivers.
Collapse
Affiliation(s)
- Lisa M Christian
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA; The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Janice K Kiecolt-Glaser
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Steve W Cole
- Departments of Psychiatry and Biobehavioral Sciences and Medicine, Division of Hematology-Oncology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Christin E Burd
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA; Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Annelise A Madison
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Veteran's Affairs Boston Healthcare System, Boston, MA 02130, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Stephanie J Wilson
- Department of Psychology, Southern Methodist University, Dallas, TX 75206, USA
| | - Ashley E Rosko
- Division of Hematology, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
22
|
Tsushima H, Tada H, Asai A, Hirose M, Hosoyama T, Watanabe A, Murakami T, Sugimoto M. Roles of pigment epithelium-derived factor in exercise-induced suppression of senescence and its impact on lung pathology in mice. Aging (Albany NY) 2024; 16:10670-10693. [PMID: 38954512 PMCID: PMC11272117 DOI: 10.18632/aging.205976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Senescent cells contribute to tissue aging and underlie the pathology of chronic diseases. The benefits of eliminating senescent cells have been demonstrated in several disease models, and the efficacy of senolytic drugs is currently being tested in humans. Exercise training has been shown to reduce cellular senescence in several tissues; however, the mechanisms responsible remain unclear. We found that myocyte-derived factors significantly extended the replicative lifespan of fibroblasts, suggesting that myokines mediate the anti-senescence effects of exercise. A number of proteins within myocyte-derived factors were identified by mass spectrometry. Among these, pigment epithelium-derived factor (PEDF) exerted inhibitory effects on cellular senescence. Eight weeks of voluntary running increased Pedf levels in skeletal muscles and suppressed senescence markers in the lungs. The administration of PEDF reduced senescence markers in multiple tissues and attenuated the decline in respiratory function in the pulmonary emphysema mouse model. We also showed that blood levels of PEDF inversely correlated with the severity of COPD in patients. Collectively, these results strongly suggest that PEDF contributes to the beneficial effects of exercise, potentially suppressing cellular senescence and its associated pathologies.
Collapse
Affiliation(s)
- Hiromichi Tsushima
- Laboratory of Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Hirobumi Tada
- Department of Nutrition, Shigakkan University, Aichi 474-8651, Japan
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Azusa Asai
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Mikako Hirose
- Laboratory of Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Tohru Hosoyama
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Atsushi Watanabe
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Taro Murakami
- Department of Nutrition, Shigakkan University, Aichi 474-8651, Japan
| | - Masataka Sugimoto
- Laboratory of Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| |
Collapse
|
23
|
Ealey KN, Togo J, Lee JH, Patel Y, Kim JR, Park SY, Sung HK. Intermittent fasting promotes rejuvenation of immunosenescent phenotypes in aged adipose tissue. GeroScience 2024; 46:3457-3470. [PMID: 38379117 PMCID: PMC11009208 DOI: 10.1007/s11357-024-01093-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/03/2024] [Indexed: 02/22/2024] Open
Abstract
The aging of white adipose tissue (WAT) involves senescence of adipose stem and progenitor cells (ASPCs) and dysregulation of immune cell populations, serving as a major driver of age-associated adipose dysfunction and metabolic diseases. Conversely, the elimination of senescent ASPCs is associated with improvements in overall health. Intermittent fasting (IF), a dietary intervention that incorporates periodic cycles of fasting and refeeding, has been reported to promote weight loss and fat mass reduction and improve glucose and insulin homeostasis in both murine and human studies. While previous studies have assessed the effects of IF on obesity-associated metabolic dysfunction, few studies have examined the aging-specific changes to ASPCs and immune cell populations in WAT. Here, we show that IF in 18-20-month-old mice reduced senescent phenotypes of ASPCs and restored their adipogenic potential. Intriguingly, IF-treated mice exhibited an increase in adipose eosinophils, which has been reported to be associated with improved WAT homeostasis and immunological fitness in aged mice. The observed cellular and metabolic changes suggest that IF may be a feasible lifestyle regimen to reduce cellular senescence which could result in attenuation of downstream aging-induced WAT dysfunction and metabolic diseases.
Collapse
Affiliation(s)
- Kafi N Ealey
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jacques Togo
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ju Hee Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Yash Patel
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jae-Ryong Kim
- Department of Biochemistry, Yeungnam University, Daegu, 42415, Republic of Korea.
- Senotherapy-based Metabolic Disease Control Research Center, Yeungnam University, Daegu, 42415, Republic of Korea.
| | - So-Young Park
- Senotherapy-based Metabolic Disease Control Research Center, Yeungnam University, Daegu, 42415, Republic of Korea.
- Department of Physiology, Yeungnam University, Daegu, 42415, Republic of Korea.
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Balasubramanian P, Kiss T, Gulej R, Nyul Toth A, Tarantini S, Yabluchanskiy A, Ungvari Z, Csiszar A. Accelerated Aging Induced by an Unhealthy High-Fat Diet: Initial Evidence for the Role of Nrf2 Deficiency and Impaired Stress Resilience in Cellular Senescence. Nutrients 2024; 16:952. [PMID: 38612986 PMCID: PMC11013792 DOI: 10.3390/nu16070952] [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: 11/22/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
High-fat diets (HFDs) have pervaded modern dietary habits, characterized by their excessive saturated fat content and low nutritional value. Epidemiological studies have compellingly linked HFD consumption to obesity and the development of type 2 diabetes mellitus. Moreover, the synergistic interplay of HFD, obesity, and diabetes expedites the aging process and prematurely fosters age-related diseases. However, the underlying mechanisms driving these associations remain enigmatic. One of the most conspicuous hallmarks of aging is the accumulation of highly inflammatory senescent cells, with mounting evidence implicating increased cellular senescence in the pathogenesis of age-related diseases. Our hypothesis posits that HFD consumption amplifies senescence burden across multiple organs. To scrutinize this hypothesis, we subjected mice to a 6-month HFD regimen, assessing senescence biomarker expression in the liver, white adipose tissue, and the brain. Aging is intrinsically linked to impaired cellular stress resilience, driven by dysfunction in Nrf2-mediated cytoprotective pathways that safeguard cells against oxidative stress-induced senescence. To ascertain whether Nrf2-mediated pathways shield against senescence induction in response to HFD consumption, we explored senescence burden in a novel model of aging: Nrf2-deficient (Nrf2+/-) mice, emulating the aging phenotype. Our initial findings unveiled significant Nrf2 dysfunction in Nrf2+/- mice, mirroring aging-related alterations. HFD led to substantial obesity, hyperglycemia, and impaired insulin sensitivity in both Nrf2+/- and Nrf2+/+ mice. In control mice, HFD primarily heightened senescence burden in white adipose tissue, evidenced by increased Cdkn2a senescence biomarker expression. In Nrf2+/- mice, HFD elicited a significant surge in senescence burden across the liver, white adipose tissue, and the brain. We postulate that HFD-induced augmentation of senescence burden may be a pivotal contributor to accelerated organismal aging and the premature onset of age-related diseases.
Collapse
Affiliation(s)
- Priya Balasubramanian
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tamas Kiss
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- International Training Program in Geroscience, First Department of Pediatrics, Semmelweis University, 1089 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 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Adam Nyul Toth
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
25
|
Wang M, Yang M, Liang S, Wang N, Wang Y, Sambou ML, Qin N, Zhu M, Wang C, Jiang Y, Dai J. Association between sleep traits and biological aging risk: a Mendelian randomization study based on 157 227 cases and 179 332 controls. Sleep 2024; 47:zsad299. [PMID: 37982786 DOI: 10.1093/sleep/zsad299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/23/2023] [Indexed: 11/21/2023] Open
Abstract
STUDY OBJECTIVES To investigate whether sleep traits are associated with the risk of biological aging using a case-control design with Mendelian randomization (MR) analyses. METHODS We studied 336 559 participants in the UK Biobank cohort, including 157 227 cases of accelerated biological aging and 179 332 controls. PhenoAge, derived from clinical traits, estimated biological ages, and the discrepancies from chronological age were defined as age accelerations (PhenoAgeAccel). Sleep behaviors were assessed with a standardized questionnaire. propensity score matching matched control participants to age-accelerated participants, and a conditional multivariable logistic regression model estimated odds ratio (OR) and 95% confidence intervals (95% CI). Causal relationships between sleep traits and PhenoAgeAccel were explored using linear and nonlinear MR methods. RESULTS A U-shaped association was found between sleep duration and PhenoAgeAccel risk. Short sleepers had a 7% higher risk (OR = 1.07; 95% CI: 1.03 to 1.11), while long sleepers had an 18% higher risk (OR = 1.18; 95% CI: 1.15 to 1.22), compared to normal sleepers (6-8 hours/day). Evening chronotype was linked to higher PhenoAgeAccel risk than morning chronotype (OR = 1.14; 95% CI: 1.10 to 1.18), while no significant associations were found for insomnia or snoring. Morning chronotype had a protective effect on PhenoAgeAccel risk (OR = 0.87, 95% CI: 0.79 to 0.95) per linear MR analysis. Genetically predicted sleep duration showed a U-shaped relationship with PhenoAgeAccel, suggesting a nonlinear association (pnonlinear < 0.001). CONCLUSIONS The study suggests that improving sleep can slow biological aging, highlighting the importance of optimizing sleep as an intervention to mitigate aging's adverse effects.
Collapse
Affiliation(s)
- Mei Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Meiqi Yang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shuang Liang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Nanxi Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yifan Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Muhammed Lamin Sambou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Na Qin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Genomic Science and Precision Medicine Institute, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Genomic Science and Precision Medicine Institute, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Cheng Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Genomic Science and Precision Medicine Institute, Gusu School, Nanjing Medical University, Nanjing 211166, China
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Yue Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Genomic Science and Precision Medicine Institute, Gusu School, Nanjing Medical University, Nanjing 211166, China
- Nanjing Yike Population Health Research Institute, Nanjing 211166, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Genomic Science and Precision Medicine Institute, Gusu School, Nanjing Medical University, Nanjing 211166, China
- Nanjing Yike Population Health Research Institute, Nanjing 211166, China
| |
Collapse
|
26
|
Fielding RA, Atkinson EJ, Aversa Z, White TA, Heeren AA, Mielke MM, Cummings SR, Pahor M, Leeuwenburgh C, LeBrasseur NK. Biomarkers of Cellular Senescence Predict the Onset of Mobility Disability and Are Reduced by Physical Activity in Older Adults. J Gerontol A Biol Sci Med Sci 2024; 79:glad257. [PMID: 37948612 PMCID: PMC10851672 DOI: 10.1093/gerona/glad257] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Indexed: 11/12/2023] Open
Abstract
Studies in mice and cross-sectional studies in humans support the premise that cellular senescence is a contributing mechanism to age-associated deficits in physical function. We tested the hypotheses that circulating proteins secreted by senescent cells are (i) associated with the incidence of major mobility disability (MMD), the development of persistent mobility disability (PMMD), and decrements in physical functioning in older adults, and (ii) influenced by physical activity (PA). Using samples and data obtained longitudinally from the Lifestyle Interventions in Elders Study clinical trial, we measured a panel of 27 proteins secreted by senescent cells. Among 1 377 women and men randomized to either a structured PA intervention or a healthy aging (HA) intervention, we observed significant associations between several senescence biomarkers, most distinctly vascular endothelial growth factor A (VEGFA), tumor necrosis factor receptor 1 (TNFR1), and matrix metallopeptidase 7 (MMP7), and the onset of both MMD and PMMD. Moreover, VEGFA, GDF15, osteopontin, and other senescence biomarkers were associated with reductions in short physical performance battery scores. The change in senescence biomarkers did not differ between PA and HA participants. In the whole cohort, higher levels of PA were associated with significantly greater reductions in 10 senescence-related proteins at 12 and/or 24 months. These data reinforce cellular senescence as a contributing mechanism of age-associated functional decline and the potential for PA to attenuate this hallmark of aging. Clinical Trials Registration Number: NCT01072500.
Collapse
Affiliation(s)
- Roger A Fielding
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Elizabeth J Atkinson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas A White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Amanda A Heeren
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Steven R Cummings
- Departments of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- Research Institute, California Pacific Medical Center, San Francisco, California, USA
| | - Marco Pahor
- Institute on Aging, University of Florida, Gainesville, Florida, USA
| | | | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
27
|
Nakagawa S, Fukui-Miyazaki A, Yoshida T, Ishii Y, Murata E, Taniguchi K, Ishizu A, Kasahara M, Tomaru U. Decreased Proteasomal Function Exacerbates Endoplasmic Reticulum Stress-Induced Chronic Inflammation in Obese Adipose Tissue. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00076-2. [PMID: 38423355 DOI: 10.1016/j.ajpath.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/24/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
Low-grade chronic inflammation contributes to both aging and the pathogenesis of age-related diseases. White adipose tissue (WAT) in obese individuals exhibits chronic inflammation, which is associated with obesity-related disorders. Aging exacerbates obesity-related inflammation in WAT; however, the molecular mechanisms underlying chronic inflammation and its exacerbation by aging remain unclear. Age-related decline in activity of the proteasome, a multisubunit proteolytic complex, has been implicated in age-related diseases. This study employed a mouse model with decreased proteasomal function that exhibits age-related phenotypes to investigate the impact of adipocyte senescence on WAT inflammation. Transgenic mice expressing proteasomal subunit β5t with weak chymotrypsin-like activity experience reduced lifespan and develop age-related phenotypes. Mice fed with a high-fat diet and experiencing proteasomal dysfunction exhibited increased WAT inflammation, increased infiltration of proinflammatory M1-like macrophages, and increased proinflammatory adipocytokine-like monocyte chemoattractant protein-1, plasminogen activator inhibitor-1, and tumor necrosis factor-α, which are all associated with activation of endoplasmic reticulum (ER) stress-related pathways. Impaired proteasomal activity also activated ER stress-related molecules and induced expression of proinflammatory adipocytokines in adipocyte-like cells differentiated from 3T3-L1 cells. Collective evidence suggests that impaired proteasomal activity increases ER stress and that subsequent inflammatory pathways play pivotal roles in WAT inflammation. Because proteasomal function declines with age, age-related proteasome impairment may be involved in obesity-related inflammation among elderly individuals.
Collapse
Affiliation(s)
- Shimpei Nakagawa
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Aya Fukui-Miyazaki
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Takuma Yoshida
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasushi Ishii
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Eri Murata
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Department of Fundamental Nursing, School of Nursing, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Koji Taniguchi
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Masanori Kasahara
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan; Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan.
| |
Collapse
|
28
|
Englund DA, Jolliffe AM, Hanson GJ, Aversa Z, Zhang X, Jiang X, White TA, Zhang L, Monroe DG, Robbins PD, Niedernhofer LJ, Kamenecka TM, Khosla S, LeBrasseur NK. Senotherapeutic drug treatment ameliorates chemotherapy-induced cachexia. JCI Insight 2024; 9:e169512. [PMID: 38051584 PMCID: PMC10906225 DOI: 10.1172/jci.insight.169512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023] Open
Abstract
Cachexia is a debilitating skeletal muscle wasting condition for which we currently lack effective treatments. In the context of cancer, certain chemotherapeutics cause DNA damage and cellular senescence. Senescent cells exhibit chronic activation of the transcription factor NF-κB, a known mediator of the proinflammatory senescence-associated secretory phenotype (SASP) and skeletal muscle atrophy. Thus, targeting NF-κB represents a logical therapeutic strategy to alleviate unintended consequences of genotoxic drugs. Herein, we show that treatment with the IKK/NF-κB inhibitor SR12343 during a course of chemotherapy reduces markers of cellular senescence and the SASP in liver, skeletal muscle, and circulation and, correspondingly, attenuates features of skeletal muscle pathology. Lastly, we demonstrate that SR12343 mitigates chemotherapy-induced reductions in body weight, lean mass, fat mass, and muscle strength. These findings support senescent cells as a promising druggable target to counteract the SASP and skeletal muscle wasting in the context of chemotherapy.
Collapse
Affiliation(s)
- Davis A. Englund
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Alyssa M. Jolliffe
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Gabriel J. Hanson
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Xinyi Jiang
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas A. White
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Lei Zhang
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - David G. Monroe
- Robert and Arlene Kogod Center on Aging, and
- Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, and
- Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
- Paul F. Glenn Center for the Biology of Aging at Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
29
|
Justice JN, Leng XI, LeBrasseur NK, Tchkonia T, Kirkland JL, Mitin N, Liu Y, Kritchevsky SB, Nicklas BJ, Ding J. Caloric Restriction Intervention Alters Specific Circulating Biomarkers of the Senescence-Associated Secretome in Middle-Aged and Older Adults With Obesity and Prediabetes in an 18-Week Randomized Controlled Trial. J Gerontol A Biol Sci Med Sci 2024; 79:glad214. [PMID: 37738560 PMCID: PMC10733170 DOI: 10.1093/gerona/glad214] [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: 03/28/2023] [Indexed: 09/24/2023] Open
Abstract
Cellular senescence is a biological aging process that is exacerbated by obesity and leads to inflammation and age- and obesogenic-driven chronic diseases including type 2 diabetes. Caloric restriction (CR) may improve metabolic function in part by reducing cellular senescence and the pro-inflammatory senescence-associated phenotype (SASP). We conducted an ancillary investigation of an 18-week randomized controlled trial (RCT) of CR (n = 31) or Control (n = 27) in 58 middle-aged/older adults (57.6 ± 5.8 years; 75% Women) with obesity and prediabetes. We measured mRNA expression of select senescence and apoptosis genes in blood CD3 + T cells (qRT-PCR) and a panel of 25 plasma SASP proteins (Luminex/multiplex; ELISA). Participants randomized to CR lost -10.8 ± 0.9 kg (-11.3% ± 5.4%) over 18 weeks compared with +0.5 ± 0.9 kg (+0.03% ± 3.5%) in Control group. T-cell expression of senescence biomarkers, p16INK4a and p21CIP1/WAF1, and apoptosis markers, BCL2L1 and BAK1, was not different between CR and Control groups in age, race, and sex-adjusted mixed models (p > .05, all). Iterative principal axis factor analysis was used to develop composite SASP Factors, and the Factors comprising TNFRI, TNFRII, uPAR, MMP1, GDF15, OPN, Fas, and MPO were significantly altered with CR intervention (age, sex, race-adjusted mixed model time × treatment F = 4.17, p ≤ .05) and associated with the degree of weight loss (R2 = 0.12, p ≤ .05). Our study provides evidence from an RCT that specific circulating biomarkers of senescent cell burden are changed by CR in middle-aged and older adults with obesity and prediabetes. Future studies compare tissue and circulating levels of p16INK4a and pro-inflammatory SASP biomarkers in other populations, and interventions.
Collapse
Affiliation(s)
- Jamie N Justice
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Xiaoyan I Leng
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Natalia Mitin
- Sapere Bio, Triangle Research Park, North Carolina, USA
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Barbara J Nicklas
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jingzhong Ding
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
30
|
Vangay P, Ward T, Lucas S, Beura LK, Sabas D, Abramson M, Till L, Hoops SL, Kashyap P, Hunter RC, Masopust D, Knights D. Industrialized human gut microbiota increases CD8+ T cells and mucus thickness in humanized mouse gut. Gut Microbes 2023; 15:2266627. [PMID: 37853762 PMCID: PMC10588527 DOI: 10.1080/19490976.2023.2266627] [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: 01/23/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023] Open
Abstract
Immigration to a highly industrialized nation has been associated with metabolic disease and simultaneous shifts in microbiota composition, but the underlying mechanisms are challenging to test in human studies. Here, we conducted a pilot study to assess the differential effects of human gut microbiota collected from the United States (US) and rural Thailand on the murine gut mucosa and immune system. Colonization of germ-free mice with microbiota from US individuals resulted in an increased accumulation of innate-like CD8 T cells in the small intestine lamina propria and intra-epithelial compartments when compared to colonization with microbiota from Thai individuals. Both TCRγδ and CD8αα T cells showed a marked increase in mice receiving Western microbiota and, interestingly, this phenotype was also associated with an increase in intestinal mucus thickness. Serendipitously, an accidentally infected group of mice corroborated this association between elevated inflammatory response and increased mucus thickness. These results suggest that Western-associated human gut microbes contribute to a pro-inflammatory immune response.
Collapse
Affiliation(s)
- Pajau Vangay
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, MN, USA
| | - Tonya Ward
- BioTechnology Institute, University of Minnesota, Minneapolis, MN, USA
| | - Sarah Lucas
- Department of Biology, Syracuse University, Syracuse, NY, USA
| | - Lalit K. Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Dominique Sabas
- Department of Food Science and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Max Abramson
- Department of Neuroscience, Macalester College, St. Paul, MN, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lisa Till
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Susan L. Hoops
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Purna Kashyap
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ryan C. Hunter
- Department of Biology, Syracuse University, Syracuse, NY, USA
| | - David Masopust
- Department of Biology, Syracuse University, Syracuse, NY, USA
| | - Dan Knights
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, MN, USA
- BioTechnology Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
31
|
Christian LM, Wilson SJ, Madison AA, Prakash RS, Burd CE, Rosko AE, Kiecolt-Glaser JK. Understanding the health effects of caregiving stress: New directions in molecular aging. Ageing Res Rev 2023; 92:102096. [PMID: 37898293 PMCID: PMC10824392 DOI: 10.1016/j.arr.2023.102096] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/11/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Dementia caregiving has been linked to multiple health risks, including infectious illness, depression, anxiety, immune dysregulation, weakened vaccine responses, slow wound healing, hypertension, cardiovascular disease, metabolic syndrome, diabetes, frailty, cognitive decline, and reduced structural and functional integrity of the brain. The sustained overproduction of proinflammatory cytokines is a key pathway behind many of these risks. However, contrasting findings suggest that some forms of caregiving may have beneficial effects, such as maintaining caregivers' health and providing a sense of meaning and purpose which, in turn, may contribute to lower rates of functional decline and mortality. The current review synthesizes these disparate literatures, identifies methodological sources of discrepancy, and integrates caregiver research with work on aging biomarkers to propose a research agenda that traces the mechanistic pathways of caregivers' health trajectories with a focus on the unique stressors facing spousal caregivers as compared to other informal caregivers. Combined with a focus on psychosocial moderators and mechanisms, studies using state-of-the-art molecular aging biomarkers such as telomere length, p16INK4a, and epigenetic age could help to reconcile mixed literature on caregiving's sequelae by determining whether and under what conditions caregiving-related experiences contribute to faster aging, in part through inflammatory biology. The biomarkers predict morbidity and mortality, and each contributes non-redundant information about age-related molecular changes -together painting a more complete picture of biological aging. Indeed, assessing changes in these biopsychosocial mechanisms over time would help to clarify the dynamic relationships between caregiving experiences, psychological states, immune function, and aging.
Collapse
Affiliation(s)
- Lisa M Christian
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Stephanie J Wilson
- Department of Psychology, Southern Methodist University, University Park, TX, USA
| | - Annelise A Madison
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Ruchika S Prakash
- Department of Psychology, The Ohio State University, Columbus, OH, USA; Center for Cognitive and Behavioral Brain Imaging, Ohio State University, Columbus, OH, USA
| | - Christin E Burd
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Ashley E Rosko
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Janice K Kiecolt-Glaser
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
32
|
Takeda R, Tabuchi A, Nonaka Y, Kano R, Sudo M, Kano Y, Hoshino D. Cmah deficiency blunts cellular senescence in adipose tissues and improves whole-body glucose metabolism in aged mice. Geriatr Gerontol Int 2023; 23:958-964. [PMID: 37968438 DOI: 10.1111/ggi.14732] [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: 06/01/2023] [Revised: 10/03/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023]
Abstract
AIM Cytidine monophosphate-N-acetylneuraminic acid (Neu5Ac) hydroxylase (Cmah) is an enzyme, which converts Neu5Ac to the sialic acid Neu5Gc. Neu5Gc is thought to increase inflammatory cytokines, which are, in part, produced in senescent cells of adipose tissues. Cellular senescence in adipose tissues induces whole-body aging and impaired glucose metabolism. Therefore, we hypothesized that Cmah deficiency would prevent cellular senescence in adipose tissues and impaired glucose metabolism. METHODS Wild-type (WT) and Cmah knockout (KO) mice aged 24-25 months were used. Whole-body metabolism was assessed using a metabolic gas analysis system. We measured blood glucose and insulin concentrations after oral glucose administration. The size of the lipid droplets in the liver was quantified. Markers of cellular senescence and senescence-associated secretory phenotypes were measured in adipose tissues. RESULTS Cmah KO had significantly increased VO2 and energy expenditure (P < 0.01). Unlike glucose, the insulin concentration after oral glucose administration was significantly lower in the Cmah KO group than in the WT group (P < 0.001). Lipid droplets in the liver were significantly lower in the Cmah KO group than in the WT group (P < 0.05). The markers of cellular senescence and senescence-associated secretory phenotypes in the adipose tissues were significantly lower in the Cmah KO group than in the WT group (P < 0.05). CONCLUSIONS Cmah deficiency blunted cellular senescence in adipose tissues and improved whole-body glucose metabolism. These characteristics in aged Cmah KO mice might be associated with higher energy expenditure. Geriatr Gerontol Int 2023; 23: 958-964.
Collapse
Affiliation(s)
- Reo Takeda
- Department of Engineering Science, The University of Electro-communications, Tokyo, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Ayaka Tabuchi
- Department of Engineering Science, The University of Electro-communications, Tokyo, Japan
| | - Yudai Nonaka
- Institute of Liberal Arts and Sciences, Kanazawa University, Ishikawa, Japan
| | - Ryotaro Kano
- Department of Engineering Science, The University of Electro-communications, Tokyo, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Mizuki Sudo
- Physical Fitness Research Institute, Meiji Yasuda Life Foundation of Health and Welfare, Tokyo, Japan
| | - Yutaka Kano
- Department of Engineering Science, The University of Electro-communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Tokyo, Japan
| | - Daisuke Hoshino
- Department of Engineering Science, The University of Electro-communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
33
|
Chin T, Lee XE, Ng PY, Lee Y, Dreesen O. The role of cellular senescence in skin aging and age-related skin pathologies. Front Physiol 2023; 14:1297637. [PMID: 38074322 PMCID: PMC10703490 DOI: 10.3389/fphys.2023.1297637] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2024] Open
Abstract
Aging is the result of a gradual functional decline at the cellular, and ultimately, organismal level, resulting in an increased risk of developing a variety of chronic illnesses, such as cardiovascular disease, stroke, cancer and diabetes. The skin is the largest organ of the human body, and the site where signs of aging are most visible. These signs include thin and dry skin, sagging, loss of elasticity, wrinkles, as well as aberrant pigmentation. The appearance of these features is accelerated by exposure to extrinsic factors such as ultraviolet (UV) radiation or pollution, as well as intrinsic factors including time, genetics, and hormonal changes. At the cellular level, aging is associated with impaired proteostasis and an accumulation of macromolecular damage, genomic instability, chromatin reorganization, telomere shortening, remodelling of the nuclear lamina, proliferation defects and premature senescence. Cellular senescence is a state of permanent growth arrest and a key hallmark of aging in many tissues. Due to their inability to proliferate, senescent cells no longer contribute to tissue repair or regeneration. Moreover, senescent cells impair tissue homeostasis, promote inflammation and extracellular matrix (ECM) degradation by secreting molecules collectively known as the "senescence-associated secretory phenotype" (SASP). Senescence can be triggered by a number of different stimuli such as telomere shortening, oncogene expression, or persistent activation of DNA damage checkpoints. As a result, these cells accumulate in aging tissues, including human skin. In this review, we focus on the role of cellular senescence during skin aging and the development of age-related skin pathologies, and discuss potential strategies to rejuvenate aged skin.
Collapse
Affiliation(s)
- Toby Chin
- Lee Kong Chiang School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Xin Er Lee
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei Yi Ng
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yaelim Lee
- Mechanobiology Institute, National University of Singapore, T-Lab, Singapore, Singapore
| | - Oliver Dreesen
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, T-Lab, Singapore, Singapore
| |
Collapse
|
34
|
Zeng L, Chen L, Gao F, Li J, Song Y, Wei L, Qu N, Li Y, Jiang H. The Comparation of Renal Anti-Senescence Effects and Blood Metabolites between Dapagliflozin and Metformin in Non-Diabetes Environment. Adv Biol (Weinh) 2023; 7:e2300199. [PMID: 37688360 DOI: 10.1002/adbi.202300199] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/10/2023] [Indexed: 09/10/2023]
Abstract
Delaying kidney senescence process will benefit renal physiologic conditions, and prompt the kidney recovering from different pathological states. The renal anti-senescence effects of sodium-glucose cotransporter-2 inhibitors (SGLT2i) and metformin have been proven in diabetic settings, but the roles of each one and combination of two drugs in natural kidney aging process remain undefined and deserve further research. Senescence-accelerated mouse prone 8 (SAMP8) were orally administered dapagliflozin, metformin, and a combination of them for 16 weeks. Dapagliflozin exhibits better effects than metformin in lowering senescence related markers, and the combination therapy shows the best results. In vitro experiments demonstrate the same results that the combination of dapagliflozin and metformin can exert a better anti-senescence effect. Blood metabolites detection in vivo shows dapagliflozin mainly leads to the change of blood metabolites enriched in choline metabolism, and metformin tends to induce change of blood metabolites enriched in purine metabolism. In conclusion, the results suggest dapagliflozin may have a better renal anti-senescence effect than metformin in non-diabetes environment, and the combination of the two drugs can strengthen the effect. The two drugs can lead to different blood metabolites alteration, which may lead to different systemic effects.
Collapse
Affiliation(s)
- Lu Zeng
- Department of Critical Care Nephrology and Blood Purification, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| | - Lei Chen
- Department of Critical Care Nephrology and Blood Purification, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| | - Fanfan Gao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| | - Jie Li
- Department of Nephrology, Henan Provincial people's hospital, Henan, 450003, China
| | - Yangyang Song
- Department of Critical Care Nephrology and Blood Purification, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| | - Limin Wei
- Department of Critical Care Nephrology and Blood Purification, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| | - Ning Qu
- Department of Medical Examination, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| | - Yan Li
- Department of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| | - Hongli Jiang
- Department of Critical Care Nephrology and Blood Purification, the First Affiliated Hospital of Xi'an Jiaotong University, Shannxi, 710061, China
| |
Collapse
|
35
|
Marcozzi S, Bigossi G, Giuliani ME, Lai G, Giacconi R, Piacenza F, Malavolta M. Spreading Senescent Cells' Burden and Emerging Therapeutic Targets for Frailty. Cells 2023; 12:2287. [PMID: 37759509 PMCID: PMC10528263 DOI: 10.3390/cells12182287] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The spreading of senescent cells' burden holds profound implications for frailty, prompting the exploration of novel therapeutic targets. In this perspective review, we delve into the intricate mechanisms underlying senescent cell spreading, its implications for frailty, and its therapeutic development. We have focused our attention on the emerging age-related biological factors, such as microbiome and virome alterations, elucidating their significant contribution to the loss of control over the accumulation rate of senescent cells, particularly affecting key frailty domains, the musculoskeletal system and cerebral functions. We believe that gaining an understanding of these mechanisms could not only aid in elucidating the involvement of cellular senescence in frailty but also offer diverse therapeutic possibilities, potentially advancing the future development of tailored interventions for these highly diverse patients.
Collapse
Affiliation(s)
- Serena Marcozzi
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121 Ancona, Italy; (S.M.); (G.B.); (M.E.G.); (R.G.); (F.P.)
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy
| | - Giorgia Bigossi
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121 Ancona, Italy; (S.M.); (G.B.); (M.E.G.); (R.G.); (F.P.)
| | - Maria Elisa Giuliani
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121 Ancona, Italy; (S.M.); (G.B.); (M.E.G.); (R.G.); (F.P.)
| | - Giovanni Lai
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121 Ancona, Italy; (S.M.); (G.B.); (M.E.G.); (R.G.); (F.P.)
| | - Robertina Giacconi
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121 Ancona, Italy; (S.M.); (G.B.); (M.E.G.); (R.G.); (F.P.)
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121 Ancona, Italy; (S.M.); (G.B.); (M.E.G.); (R.G.); (F.P.)
| | - Marco Malavolta
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121 Ancona, Italy; (S.M.); (G.B.); (M.E.G.); (R.G.); (F.P.)
| |
Collapse
|
36
|
Sandstedt M, Chung RWS, Skoglund C, Lundberg AK, Östgren CJ, Ernerudh J, Jonasson L. Complete fatty degeneration of thymus associates with male sex, obesity and loss of circulating naïve CD8 + T cells in a Swedish middle-aged population. Immun Ageing 2023; 20:45. [PMID: 37653480 PMCID: PMC10470174 DOI: 10.1186/s12979-023-00371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Fatty degeneration of thymus (or thymus involution) has long been considered a normal ageing process. However, there is emerging evidence that thymic involution is linked to T cell aging, chronic inflammation and increased morbidity. Other factors, aside from chronological age, have been proposed to affect the involution rate. In the present study, we investigated the imaging characteristics of thymus on computed tomography (CT) in a Swedish middle-aged population. The major aims were to establish the prevalence of fatty degeneration of thymus and to determine its associations with demographic, lifestyle and clinical factors, as well as inflammation, T cell differentiation and thymic output. RESULTS In total, 1 048 randomly invited individuals (aged 50-64 years, 49% females) were included and thoroughly characterized. CT evaluation of thymus included measurements of attenuation, size and a 4-point scoring system, with scale 0-3 based on the ratio of fat and soft tissue. A majority, 615 (59%) showed complete fatty degeneration, 259 (25%) predominantly fatty attenuation, 105 (10%) half fatty and half soft-tissue attenuation, while 69 (6.6%) presented with a solid thymic gland with predominantly soft-tissue attenuation. Age, male sex, high BMI, abdominal obesity and low dietary intake of fiber were independently associated with complete fatty degeneration of thymus. Also, fatty degeneration of thymus as well as low CT attenuation values were independently related to lower proportion of naïve CD8+ T cells, which in turn was related to lower thymic output, assessed by T-cell receptor excision circle (TREC) levels. CONCLUSION Among Swedish middle-aged subjects, nearly two-thirds showed complete fatty degeneration of thymus on CT. This was linked to depletion of naïve CD8+ T cells indicating that CT scans of thymus might be used to estimate immunological aging. Furthermore, our findings support the intriguing concept that obesity as well as low fiber intake contribute to immunological aging, thereby raising the possibility of preventive strategies.
Collapse
Affiliation(s)
- Mårten Sandstedt
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Radiology and Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Rosanna W S Chung
- Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Camilla Skoglund
- Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Anna K Lundberg
- Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Carl Johan Östgren
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine and Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Lena Jonasson
- Department of Cardiology and Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
37
|
Iwasaki K, Lalani B, Kahng J, Carapeto P, Sanjines S, Hela F, Abarca C, Tsuji T, Darcy J, Bartke A, Tseng YH, Kulkarni RN, Aguayo-Mazzucato C. Decreased IGF1R attenuates senescence and improves function in pancreatic β-cells. Front Endocrinol (Lausanne) 2023; 14:1203534. [PMID: 37441495 PMCID: PMC10335398 DOI: 10.3389/fendo.2023.1203534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/02/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction The enhanced β-cell senescence that accompanies insulin resistance and aging contributes to cellular dysfunction and loss of transcriptional identity leading to type 2 diabetes (T2D). While senescence is among the 12 recognized hallmarks of aging, its relation to other hallmarks including altered nutrient sensing (insulin/IGF1 pathway) in β-cells is not fully understood. We previously reported that an increased expression of IGF1R in mouse and human β-cells is a marker of older β-cells; however, its contribution to age-related dysfunction and cellular senescence remains to be determined. Methods In this study, we explored the direct role of IGF1R in β-cell function and senescence using two independent mouse models with decreased IGF1/IGF1R signaling: a) Ames Dwarf mice (Dwarf +/+), which lack growth hormone and therefore have reduced circulating levels of IGF1, and b) inducible β-cell-specific IGF1R knockdown (βIgf1rKD) mice. Results Compared to Dwarf+/- mice, Dwarf+/+ mice had lower body and pancreas weight, lower circulating IGF1 and insulin levels, and lower IGF1R and p21Cip1 protein expression in β-cells, suggesting the suppression of senescence. Adult βIgf1rKD mice showed improved glucose clearance and glucose-induced insulin secretion, accompanied by decreased p21Cip1 protein expression in β-cells. RNA-Seq of islets isolated from these βIgf1rKD mice revealed the restoration of three signaling pathways known to be downregulated by aging: sulfide oxidation, autophagy, and mTOR signaling. Additionally, deletion of IGF1R in mouse β-cells increased transcription of genes important for maintaining β-cell identity and function, such as Mafa, Nkx6.1, and Kcnj11, while decreasing senescence-related genes, such as Cdkn2a, Il1b, and Serpine 1. Decreased senescence and improved insulin-secretory function of β-cells were also evident when the βIgf1rKD mice were fed a high-fat diet (HFD; 60% kcal from fat, for 5 weeks). Discussion These results suggest that IGF1R signaling plays a causal role in aging-induced β-cell dysfunction. Our data also demonstrate a relationship between decreased IGF1R signaling and suppressed cellular senescence in pancreatic β-cells. Future studies can further our understanding of the interaction between senescence and aging, developing interventions that restore β-cell function and identity, therefore preventing the progression to T2D.
Collapse
Affiliation(s)
- Kanako Iwasaki
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
- Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Benjamin Lalani
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Jiho Kahng
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Priscila Carapeto
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Stephanie Sanjines
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Francesko Hela
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Cristian Abarca
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Justin Darcy
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Andrzej Bartke
- Department of Internal Medicine, Division of Geriatrics Research, Department of Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Rohit N. Kulkarni
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | - Cristina Aguayo-Mazzucato
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
38
|
Zhang T, He Y, Shu X, Ma X, Wu J, Du Z, Xu J, Chen N, You J, Liu Y, Li T, Wu J. Photomodulation alleviates cellular senescence of aging adipose-derived stem cells. Cell Commun Signal 2023; 21:146. [PMID: 37337219 DOI: 10.1186/s12964-023-01152-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/28/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) therapies are emerging as a promising approach to therapeutic regeneration. Therapeutic persistence and reduced functional stem cells following cell delivery remain critical hurdles for clinical investigation due to the senescence of freshly isolated cells and extensive in-vitro passage. METHODS Cultured adipose-derived stem cells (ASCs) were derived from subcutaneous white adipose tissue isolated from mice fed a normal diet. We performed senescence-associated-β-galactosidase (SA-β-gal) staining, real-time PCR, and Westernblot to evaluate the levels related to cellular senescence markers. RESULTS The mRNA expression levels of senescence markers were significantly increased in the later passage of ASCs. We show that light activation reduced the expression of senescent genes, and SA-β-Gal in all cells at passages. Moreover, the light-activated ASCs-derived exosomes decrease the expression of senescence, and SA-β-Gal in the later passage cells. We further investigated the photoreceptive effect of Opsin3 (Opn3) in light-activated ASCs. Deletion of Opn3 abolished the differences of light activation in reduced expression of senescent genes, increased Ca 2+ influx, and cAMP levels. CONCLUSIONS ASCs can undergo cellular senescence in-vitro passage. Photomodulation might be better preserved over senescence and Opn3-dependent activation in aged ASCs. Light-activated ASCs-derived exosomes could be served as e a new protective paradigm for cellular senescence in-vitro passage. Video Abstract.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Yuqian He
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Xin Shu
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Xiaoyu Ma
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Jiaqi Wu
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Zuoqin Du
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Jin Xu
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Ni Chen
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Jingcan You
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Yaofang Liu
- Department of Reproductive Technology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tian Li
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China
| | - Jianbo Wu
- Department of Pharmacology, School of Pharmacy Drug Discovery Research Center of Southwest Medical University, and Laboratory for Cardiovascular Pharmacology, Southwest Medical University, Luzhou, Sichuan, China.
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, 646000, Sichuan, China.
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
39
|
Nehme J, Altulea A, Gheorghe T, Demaria M. The effects of macronutrients metabolism on cellular and organismal aging. Biomed J 2023; 46:100585. [PMID: 36801257 PMCID: PMC10209809 DOI: 10.1016/j.bj.2023.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Evidence supports the notion that metabolic pathways are major regulators of organismal aging, and that metabolic perturbations can extend health- and lifespan. For this reason, dietary interventions and compounds perturbing metabolism are currently explored as anti-aging strategies. A common target for metabolic interventions delaying aging is cellular senescence, a state of stable growth arrest that is accompanied by various structural and functional changes including the activation of a pro-inflammatory secretome. Here, we summarize the current knowledge on the molecular and cellular events associated with carbohydrate, lipid and protein metabolism, and define how macronutrients can regulate induction or prevention of cellular senescence. We discuss how various dietary interventions can achieve prevention of disease and extension of healthy longevity by partially modulating senescence-associated phenotypes. We also emphasize the importance of developing personalized nutritional interventions that take into account the current health and age status of the individual.
Collapse
Affiliation(s)
- Jamil Nehme
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Abdullah Altulea
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Teodora Gheorghe
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Marco Demaria
- University of Groningen, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands.
| |
Collapse
|
40
|
Chen Q, Young L, Barsotti R. Mitochondria in cell senescence: A Friend or Foe? ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:35-91. [PMID: 37437984 DOI: 10.1016/bs.apcsb.2023.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Cell senescence denotes cell growth arrest in response to continuous replication or stresses damaging DNA or mitochondria. Mounting research suggests that cell senescence attributes to aging-associated failing organ function and diseases. Conversely, it participates in embryonic tissue maturation, wound healing, tissue regeneration, and tumor suppression. The acute or chronic properties and microenvironment may explain the double faces of senescence. Senescent cells display unique characteristics. In particular, its mitochondria become elongated with altered metabolomes and dynamics. Accordingly, mitochondria reform their function to produce more reactive oxygen species at the cost of low ATP production. Meanwhile, destructed mitochondrial unfolded protein responses further break the delicate proteostasis fostering mitochondrial dysfunction. Additionally, the release of mitochondrial damage-associated molecular patterns, mitochondrial Ca2+ overload, and altered NAD+ level intertwine other cellular organelle strengthening senescence. These findings further intrigue researchers to develop anti-senescence interventions. Applying mitochondrial-targeted antioxidants reduces cell senescence and mitigates aging by restoring mitochondrial function and attenuating oxidative stress. Metformin and caloric restriction also manifest senescent rescuing effects by increasing mitochondria efficiency and alleviating oxidative damage. On the other hand, Bcl2 family protein inhibitors eradicate senescent cells by inducing apoptosis to facilitate cancer chemotherapy. This review describes the different aspects of mitochondrial changes in senescence and highlights the recent progress of some anti-senescence strategies.
Collapse
Affiliation(s)
- Qian Chen
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States.
| | - Lindon Young
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Robert Barsotti
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| |
Collapse
|
41
|
Félix-Soriano E, Sáinz N, Gil-Iturbe E, Castilla-Madrigal R, Celay J, Fernández-Galilea M, Pejenaute Á, Lostao MP, Martínez-Climent JA, Moreno-Aliaga MJ. Differential remodeling of subcutaneous white and interscapular brown adipose tissue by long-term exercise training in aged obese female mice. J Physiol Biochem 2023:10.1007/s13105-023-00964-2. [PMID: 37204588 DOI: 10.1007/s13105-023-00964-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 04/26/2023] [Indexed: 05/20/2023]
Abstract
Obesity exacerbates aging-induced adipose tissue dysfunction. This study aimed to investigate the effects of long-term exercise on inguinal white adipose tissue (iWAT) and interscapular brown adipose tissue (iBAT) of aged obese mice. Two-month-old female mice received a high-fat diet for 4 months. Then, six-month-old diet-induced obese animals were allocated to sedentarism (DIO) or to a long-term treadmill training (DIOEX) up to 18 months of age. In exercised mice, iWAT depot revealed more adaptability, with an increase in the expression of fatty acid oxidation genes (Cpt1a, Acox1), and an amelioration of the inflammatory status, with a favorable modulation of pro/antiinflammatory genes and lower macrophage infiltration. Additionally, iWAT of trained animals showed an increment in the expression of mitochondrial biogenesis (Pgc1a, Tfam, Nrf1), thermogenesis (Ucp1), and beige adipocytes genes (Cd137, Tbx1). In contrast, iBAT of aged obese mice was less responsive to exercise. Indeed, although an increase in functional brown adipocytes genes and proteins (Pgc1a, Prdm16 and UCP1) was observed, few changes were found on inflammation-related and fatty acid metabolism genes. The remodeling of iWAT and iBAT depots occurred along with an improvement in the HOMA index for insulin resistance and in glucose tolerance. In conclusion, long-term exercise effectively prevented the loss of iWAT and iBAT thermogenic properties during aging and obesity. In iWAT, the long-term exercise program also reduced the inflammatory status and stimulated a fat-oxidative gene profile. These exercise-induced adipose tissue adaptations could contribute to the beneficial effects on glucose homeostasis in aged obese mice.
Collapse
Affiliation(s)
- Elisa Félix-Soriano
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Neira Sáinz
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Eva Gil-Iturbe
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Rosa Castilla-Madrigal
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Jon Celay
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, University of Navarra, Pamplona, Spain
- CIBERONC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Marta Fernández-Galilea
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Álvaro Pejenaute
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - M Pilar Lostao
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - José A Martínez-Climent
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, University of Navarra, Pamplona, Spain
- CIBERONC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - María J Moreno-Aliaga
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
42
|
Lee HJ, Yoon YS, Lee SJ. Molecular mechanisms of cellular senescence in neurodegenerative diseases. J Mol Biol 2023:168114. [PMID: 37085010 DOI: 10.1016/j.jmb.2023.168114] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Neurodegenerative diseases, such as Alzheimer's and Parkinson's, are characterized by several pathological features, including selective neuronal loss, aggregation of specific proteins, and chronic inflammation. Aging is the most critical risk factor of these disorders. However, the mechanism by which aging contributes to the pathogenesis of neurodegenerative diseases is not clearly understood. Cellular senescence is a cell state or fate in response to stimuli. It is typically associated with a series of changes in cellular phenotypes such as abnormal cellular metabolism and proteostasis, reactive oxygen species (ROS) production, and increased secretion of certain molecules via senescence-associated secretory phenotype (SASP). In this review, we discuss how cellular senescence contributes to brain aging and neurodegenerative diseases, and the relationship between protein aggregation and cellular senescence. Finally, we discuss the potential of senescence modifiers and senolytics in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- He-Jin Lee
- Department of Anatomy, Konkuk University, Seoul 05029, Korea; IBST, Konkuk University, Seoul 05029, Korea.
| | - Ye-Seul Yoon
- Department of Anatomy, Konkuk University, Seoul 05029, Korea; IBST, Konkuk University, Seoul 05029, Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, Korea; Neuramedy, Co., Ltd., Seoul, Korea.
| |
Collapse
|
43
|
A cross-talk between sestrins, chronic inflammation and cellular senescence governs the development of age-associated sarcopenia and obesity. Ageing Res Rev 2023; 86:101852. [PMID: 36642190 DOI: 10.1016/j.arr.2023.101852] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
The rapid increase in both the lifespan and proportion of older adults is accompanied by the unprecedented rise in age-associated chronic diseases, including sarcopenia and obesity. Aging is also manifested by increased susceptibility to multiple endogenous and exogenous stresses enabling such chronic conditions to develop. Among the main physiological regulators of cellular adaption to various stress stimuli, such as DNA damage, hypoxia, and oxidative stress, are sestrins (Sesns), a family of three evolutionarily conserved proteins, Sesn1, 2, and 3. Age-associated sarcopenia and obesity are characterized by two key processes: (i) accumulation of senescent cells in the skeletal muscle and adipose tissue and (ii) creation of a systemic, chronic, low-grade inflammation (SCLGI). Presumably, failed SCLGI resolution governs the development of these chronic conditions. Noteworthy, Sesns activate senolytics, which are agents that selectively eliminate senescent cells, as well as specialized pro-resolving mediators, which are factors that physiologically provide inflammation resolution. Sesns reveal clear beneficial effects in pre-clinical models of sarcopenia and obesity. Based on these observations, we propose a novel treatment strategy for age-associated sarcopenia and obesity, complementary to the conventional therapeutic modalities: Sesn activation, SCLGI resolution, and senescent cell elimination.
Collapse
|
44
|
Hsieh CC, Chang CY, Yar Lee TX, Wu J, Saovieng S, Hsieh YW, Zhu M, Huang CY, Kuo CH. Longevity, tumor, and physical vitality in rats consuming ginsenoside Rg1. J Ginseng Res 2023; 47:210-217. [PMID: 36926614 PMCID: PMC10014179 DOI: 10.1016/j.jgr.2021.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/12/2021] [Accepted: 04/18/2021] [Indexed: 10/21/2022] Open
Abstract
Background Effects of the major ginsenoside Rg1 on mammalian longevity and physical vitality are rarely reported. Purpose To examine longevity, tumor, and spontaneous locomotor activity in rats consuming Rg1. Methods A total of 138 Wistar rats were randomized into 2 groups: control (N = 69) and Rg1 (N = 69). Rg1 (0.1 mg/kg per day) were orally supplemented from 6 months of age until natural death. Spontaneous mobility was measured by video-tracking together with body composition (dual energy x-ray absorptiometry) and inflammation markers at 5, 14, 21, and 28 months of age. Results No significant differences in longevity (control: 706 days; Rg1: 651 days, p = 0.77) and tumor incidence (control: 19%; Rg1: 12%, p = 0.24) were observed between the two groups. Movement distance in the control group declined significantly by ∼60% at 21 months of age, together with decreased TNF-α (p = 0.01) and increased IL-10 (p = 0.02). However, the movement distance in the Rg1 group was maintained ∼50% above the control groups (p = 0.01) at 21 months of age with greater magnitudes of TNF-α decreases and IL-10 increases. Glucose, insulin, and body composition (bone, muscle and fat percentages) were similar for both groups during the entire observation period. Conclusion The results of the study suggest a delay age-dependent decline in physical vitality during late life by lifelong Rg1 consumption. This improvement is associated with inflammatory modulation. Significant effects of Rg1 on longevity and tumorigenesis were not observed.
Collapse
Affiliation(s)
- Chao-Chieh Hsieh
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Chiung-Yun Chang
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Tania Xu Yar Lee
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Jinfu Wu
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan.,Laboratory of Regenerative Medicine in Sports Science, School of Physical Education & Sports Science, South China Normal University, Guangzhou, China
| | - Suchada Saovieng
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan.,College of Sports Science & Technology, Mahidol University, Thailand
| | - Yu-Wen Hsieh
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Maijian Zhu
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| |
Collapse
|
45
|
Seitz-Holland J, Mulsant BH, Reynolds CF, Blumberger DM, Karp JF, Butters MA, Mendes-Silva AP, Vieira EL, Tseng G, Lenze EJ, Diniz BS. Major depression, physical health and molecular senescence markers abnormalities. NATURE. MENTAL HEALTH 2023; 1:200-209. [PMID: 39483500 PMCID: PMC11527398 DOI: 10.1038/s44220-023-00033-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/10/2023] [Indexed: 11/03/2024]
Abstract
Previous studies suggested the role of cellular senescence in late-life depression (LLD). However, it is unclear how this finding relates to common features of LLD, such as medical and cognitive problems. We applied factor analyses to an extensive battery of clinical variables in 426 individuals with LLD. Here we tested the relationship between these factors, age and sex, with an index of cellular senescence based on 22 senescence-associated secretory phenotype proteins. We found four factors: 'depression and anxiety severity', 'cognitive functioning', 'cardiovascular and cardiometabolic health' and 'blood pressure'. A higher senescence-associated secretory phenotype index was associated with poorer 'cognitive functioning' and 'cardiovascular and cardiometabolic health' but not with 'depression and anxiety severity'. These findings highlight the role of cellular senescence in poorer physical and cognitive health in LLD. They are consonant with the viewpoint that co-occurring medical burdens and their associated disabilities are part of a phenotype of accelerated ageing in LLD.
Collapse
Affiliation(s)
- Johanna Seitz-Holland
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benoit H. Mulsant
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles F. Reynolds
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daniel M. Blumberger
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jordan F. Karp
- Department of Psychiatry, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Meryl A. Butters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Erica L. Vieira
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Eric J. Lenze
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Breno S. Diniz
- UConn Center on Aging, University of Connecticut, Farmington, CT, USA
- Department of Psychiatry, UConn School of Medicine, Farmington, CT, USA
| |
Collapse
|
46
|
Nehme J, Yang D, Altulea A, Varela-Eirin M, Wang L, Hu S, Wu Y, Togo J, Niu C, Speakman JR, Demaria M. High dietary protein and fat contents exacerbate hepatic senescence and SASP in mice. FEBS J 2023; 290:1340-1347. [PMID: 34908245 DOI: 10.1111/febs.16292] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/11/2021] [Accepted: 11/22/2021] [Indexed: 01/10/2023]
Abstract
Dietary choices have a profound impact on the aging process. In addition to the total amount of energy intake, macronutrient composition influences both health and lifespan. However, the exact mechanisms by which dietary macronutrients influence onset and progression of age-associated features remain poorly understood. Cellular senescence is a state of stable growth arrest characterized by the secretion of numerous bioactive molecules with pro-inflammatory properties. Accumulation of senescent cells is considered one of the basic mechanisms of aging and an important contributor to chronic inflammation and tissue degeneration. Whether dietary macronutrients affect the accumulation and the phenotype of senescent cells with age is still unknown. Here, we show that feeding on diets with varying ratios of dietary macronutrients for 3 months has a significant effect on different senescence-associated markers in the mouse liver. High protein intake is associated with higher expression levels of the two classical senescence-associated growth arrest genes, p21 and p16. Furthermore, the expression of many pro-inflammatory secretory markers was increased in diets enriched in protein and further enhanced by increases in fat content. These results provide preliminary evidence that dietary macronutrients have a significant influence on senescence markers and merit further investigation.
Collapse
Affiliation(s)
- Jamil Nehme
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), The Netherlands.,Doctoral School of Science and Technology, Lebanese University, Hadath, Beirut, Lebanon
| | - Dengbao Yang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing Technology and Business University, China
| | - Abdullah Altulea
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), The Netherlands
| | - Marta Varela-Eirin
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), The Netherlands
| | - Lu Wang
- School of Pharmacy, Yantai University, China
| | - Sumei Hu
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing Technology and Business University, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, National Soybean Processing Industry Technology Innovation Center, Beijing Technology and Business University, China
| | - Yingga Wu
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing Technology and Business University, China
| | - Jacques Togo
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing Technology and Business University, China
| | - Chaoqun Niu
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing Technology and Business University, China
| | - John R Speakman
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing Technology and Business University, China.,Institute of Biological and Environmental Sciences, University of Aberdeen, UK
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), The Netherlands
| |
Collapse
|
47
|
Ruggiero AD, Davis MA, Davis AT, DeStephanis D, Williams AG, Vemuri R, Fanning KM, Sherrill C, Cline JM, Caudell DL, Kavanagh K. Delayed effects of radiation in adipose tissue reflect progenitor damage and not cellular senescence. GeroScience 2023; 45:507-521. [PMID: 36136223 PMCID: PMC9886706 DOI: 10.1007/s11357-022-00660-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/08/2022] [Indexed: 02/03/2023] Open
Abstract
The pathogenesis of many age-related diseases is linked to cellular senescence, a state of inflammation-inducing, irreversible cell cycle arrest. The consequences and mechanisms of age-associated cellular senescence are often studied using in vivo models of radiation exposure. However, it is unknown whether radiation induces persistent senescence, like that observed in ageing. We performed analogous studies in mice and monkeys, where young mice and rhesus macaques received sub-lethal doses of ionizing radiation and were observed for ~ 15% of their expected lifespan. Assessments of 8-hydroxy-2' -deoxyguanosine (8-OHdG), senescence-associated beta-galactosidase (SAβ-gal), and p16Ink4a and p21 were performed on mitotic and post-mitotic tissues - liver and adipose tissue - 6 months and 3 years post-exposure for the mice and monkeys, respectively. No elevations in 8-OHdG, SA-βgal staining, or p16 Ink4a or p21 gene or protein expression were found in mouse and monkey liver or adipose tissue compared to control animals. Despite no evidence of senescence, progenitor cell dysfunction persisted after radiation exposure, as indicated by lower in situ CD34+ adipose cells (p = 0.03), and deficient adipose stromal vascular cell proliferation (p < 0.05) and differentiation (p = 0.04) ex vivo. Our investigation cautions that employing radiation to study senescence-related processes should be limited to the acute post-exposure period and that stem cell damage likely underpins the dysfunction associated with delayed effects of radiation.
Collapse
Affiliation(s)
- Alistaire D Ruggiero
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Matthew A Davis
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ashley T Davis
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Darla DeStephanis
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Abigail G Williams
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Ravichandra Vemuri
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Katherine M Fanning
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Chrissy Sherrill
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - J Mark Cline
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - David L Caudell
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Kylie Kavanagh
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA.
- College of Health and Medicine, University of Tasmania, Hobart, Australia.
| |
Collapse
|
48
|
Conover CA, Bale LK. Senescence induces proteolytically-active PAPP-A secretion and association with extracellular vesicles in human pre-adipocytes. Exp Gerontol 2023; 172:112070. [PMID: 36549546 PMCID: PMC9868105 DOI: 10.1016/j.exger.2022.112070] [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] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Senescence is a cellular response to various stressors characterized by irreversible cell cycle arrest, resistance to apoptosis and expression of a senescence-associated secretory phenotype (SASP). Interestingly, studies where senescent cells were deleted in mice produced beneficial effects similar to those where the zinc metalloproteinase, PAPP-A, was deleted in mice. In this study, we investigated the effect of senescence on PAPP-A secretion and activity in primary cultures of adult human pre-adipocytes. Cultured pre-adipocytes were isolated from subcutaneous (Sub) and omental (Om) fat. Senescence was induced with low dose etoposide. PAPP-A protein was measured by an ultrasensitive PAPP-A ELISA. PAPP-A proteolytic activity was measured by a specific substrate cleavage assay. Senescence significantly increased PAPP-A levels in both Sub and Om conditioned medium (CM) 8- to 15-fold over non-senescent CM. Proteolytic activity reflected PAPP-A protein with 12- to 18-fold greater activity in senescent CM versus non-senescent CM. Furthermore, PAPP-A was found at high levels on the surface of extracellular vesicles secreted by senescent pre-adipocytes and was proteolytically active. In conclusion, we identified enzymatically active PAPP-A as a component of human pre-adipocyte SASP. This recognition warrants further investigation of PAPP-A as a new biomarker for senescence and a potential therapeutic target to control of the spread of senescence in adipose tissue.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Metabolism and Nutrition, Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA.
| | - Laurie K Bale
- Division of Endocrinology, Metabolism and Nutrition, Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
49
|
Obesity triggers tumoral senescence and renders poorly immunogenic malignancies amenable to senolysis. Proc Natl Acad Sci U S A 2023; 120:e2209973120. [PMID: 36574648 PMCID: PMC9910606 DOI: 10.1073/pnas.2209973120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Obesity is a major risk factor for cancer. Conventional thought suggests that elevated adiposity predisposes to heightened inflammatory stress and potentiates tumor growth, yet underlying mechanisms remain ill-defined. Here, we show that tumors from patients with a body mass index >35 carry a high burden of senescent cells. In mouse syngeneic tumor models, we correlated a pronounced accretion of senescent cancer cells with poorly immunogenic tumors when mice were subjected to diet-induced obesity (DIO). Highly immunogenic tumors showed lesser senescence burden suggesting immune-mediated elimination of senescent cancer cells, likely targeted as a consequence of their senescence-associated secretory phenotype. Treatment with the senolytic BH3 mimetic small molecule inhibitor ABT-263 selectively stalled tumor growth in mice with DIO to rates comparable to regular diet-fed mice. Thus, consideration of body adiposity in the selection of cancer therapy may be a critical determinant for disease outcome in poorly immunogenic malignancies.
Collapse
|
50
|
Yu Q, Walters HE, Yun MH. Induction and Characterization of Cellular Senescence in Salamanders. Methods Mol Biol 2023; 2562:135-154. [PMID: 36272072 DOI: 10.1007/978-1-0716-2659-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cellular senescence is a permanent proliferation arrest mechanism induced following the detection of genotoxic stress. Mounting evidence has causally linked the accumulation of senescent cells to a growing number of age-related pathologies in mammals. However, recent data have also highlighted senescent cells as important mediators of tissue remodeling during organismal development, tissue repair, and regeneration. As powerful model organisms for studying such processes, salamanders constitute a system in which to probe the characteristics, physiological functions, and evolutionary facets of cellular senescence. In this chapter, we outline methods for the generation, identification, and characterization of salamander senescent cells in vitro and in vivo.
Collapse
Affiliation(s)
- Qinghao Yu
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Hannah E Walters
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany.
| | - Maximina H Yun
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany.
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|