1
|
Di W, Xue C, Lin Y, Zhang W, Zhou Y. Exosome miR-152-3p derived from small intestinal epithelium modulates aging process in adipocytes. 3 Biotech 2025; 15:163. [PMID: 40375937 PMCID: PMC12075044 DOI: 10.1007/s13205-025-04346-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: 02/09/2025] [Accepted: 05/04/2025] [Indexed: 05/18/2025] Open
Abstract
Exosomes play a crucial role in facilitating intracellular communication between cells and tissues. The small intestine epithelium secretes exosomes, which is involved in various physiologic and pathologic processes. In this study, we investigated the effects of exosomal miR-152-3p derived from small intestinal epithelium on the aging process of adipocytes and its potential downstream mechanism. The exosomes derived from small intestinal epithelial cells were identified and characterized by TEM, NTA, and Western blot (WB). CCK-8 assay demonstrated the concentration-dependently increased 3T3-L1 cell viability by exosomes. PCR, Mito-Tracker red and DCFH-DA staining demonstrated the increased mtDNA content, mitochondrial activity, and the declined ROS content in 3T3-L1 adipocytes co-cultured with young exosomes. WB, PCR, β-galactosidase staining and ELISA demonstrated that the senescence was suppressed, uncoupling protein 1 (UCP1) and PPARgamma coactivator 1-alpha (PGC-1α) expression were upregulated, the levels of proinflammatory tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6) were decreased in 3T3-L1 adipocytes co-cultured with young exosomes. Luciferase reporter assay determined the binding between miR-152-3p and PGC-1α. WB and PCR manifested that miR-152-3p was lowly expressed in young exosomes and miR-152-3p could decrease PGC-1α expression and increase the expression of senescence-related genes. Moreover, ITT and GTT and H&E staining in in vivo elderly mouse model demonstrated that miR-152-3p inhibitor decreased visceral fat, improved glucose tolerance and insulin sensitivity and inhibited aging. WB and PCR suggested that miR-152-3p inhibitor enhanced PGC-1α expression, suppressed the expression of senescence-related genes and proinflammatory factors in vivo. In summary, intestinal exosomes affect the browning function of adipocytes through miR-152-3p, modulating the aging process.
Collapse
Affiliation(s)
- Wenjuan Di
- Department of Geriatrics, Division of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Cheng Xue
- Department of Geriatrics, Division of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Yunyun Lin
- Department of Geriatrics, Division of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Wenling Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Yichan Zhou
- Department of Geriatrics, Division of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
2
|
Zong Y, Han Z, Xu L, Zhang Y, Chen W, Liu T. Identification of Hub Genes and Pathways Associated with Ageing in Diabetic Encephalopathy Based on Transcriptome Analysis. Biochem Genet 2025:10.1007/s10528-025-11134-y. [PMID: 40397335 DOI: 10.1007/s10528-025-11134-y] [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: 07/31/2024] [Accepted: 05/06/2025] [Indexed: 05/22/2025]
Abstract
This study aimed to identify key genes and pathways associated with ageing in diabetic encephalopathy (DE) through transcriptome analysis and to explore their roles and mechanisms in accelerating brain ageing in diabetes. We used db/db mice to establish a model of type 2 diabetes mellitus DE. Moreover, ribonucleic acid sequencing was performed on hippocampal tissue, and differentially expressed genes (DEGs) were analysed. Ageing-related DEGs (Ag-DEGs) were identified based on the GenAge and CellAge databases. A protein-protein interaction (PPI) network of Ag-DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins database, and hub genes were identified using the Molecular Complex Detection and CytoHubba plugins of Cytoscape. Finally, immune infiltration analysis was conducted based on transcriptome data to investigate the role of immune cells in diabetic brain ageing. A total of 98 Ag-DEGs were identified, primarily involved in hypoxia, tumour necrosis factor-alpha signalling via nuclear factor kappa B, apoptosis and P53 pathways. The PPI network analysis identified 14 hub genes: HDAC1, IGF2, EGR1, BCL2, FOS, ATM, EGF, PARP1, MAPK3, APOE, SOX2, CAV1, HSPA5 and NFKBIA. These genes play significant roles in apoptosis pathways in cancer, lipid metabolism, atherosclerosis and human immunodeficiency virus-1 infection. Immune infiltration analysis revealed significant differences in the distribution of natural killer cells, resting mast cells and plasma cells within the diabetic brain. This study identified Ag-DEGs and hub genes in a DE model, revealing potential mechanisms of diabetes-accelerated brain ageing. These findings provide new insights into the pathological mechanisms of diabetic brain ageing and may offer new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Yonghua Zong
- Key Laboratory of Health-Cultivation, Ministry of Education of the People's Republic of China, Beijing University of Chinese Medicine, Xueyuan South Street, Gongchen Street, Fangshan District, Beijing, 100029, China
- Department of Tibetan Medicine, University of Tibetan Medicine, Lhasa, 850000, China
| | - Zekun Han
- Key Laboratory of Health-Cultivation, Ministry of Education of the People's Republic of China, Beijing University of Chinese Medicine, Xueyuan South Street, Gongchen Street, Fangshan District, Beijing, 100029, China
| | - Lijun Xu
- Institute of Tibetan Medicine, University of Tibetan Medicine, Lhasa, 850000, China
| | - Yanfei Zhang
- Institute of Tibetan Medicine, University of Tibetan Medicine, Lhasa, 850000, China
| | - Wanlin Chen
- Chengdu Medical College, Chengdu, 610000, China
| | - Tonghua Liu
- Key Laboratory of Health-Cultivation, Ministry of Education of the People's Republic of China, Beijing University of Chinese Medicine, Xueyuan South Street, Gongchen Street, Fangshan District, Beijing, 100029, China.
| |
Collapse
|
3
|
Zhang S, Han B, Wang X, Yuan X. Modulation of adipose tissue phenotype and longevity-related gene expression by nuclear factor E2-related factor 2 knockdown in 3T3-L1 cells. Hormones (Athens) 2025:10.1007/s42000-025-00669-9. [PMID: 40341662 DOI: 10.1007/s42000-025-00669-9] [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: 12/22/2024] [Accepted: 04/24/2025] [Indexed: 05/10/2025]
Abstract
PURPOSE This study explores the role of nuclear factor E2-related factor 2 (Nrf2) in regulating adipose tissue phenotype and its potential mechanisms for promoting aging resistance in 3T3-L1 adipocytes. The study aims to evaluate the impact of Nrf2 knockdown on adipose phenotype transformation, focusing on brown adipose tissue (BAT) and white adipose tissue (WAT) marker genes, as well as longevity-related factors. METHODS 3T3-L1 preadipocytes were differentiated into adipocytes using a standard MDI regimen. Nrf2 expression was knocked down via siRNA transfection. Gene expression was assessed using quantitative real-time PCR (qPCR), and protein levels were analyzed using Western blotting. RESULTS Nrf2 knockdown was confirmed by Western blot (p<0.001) and qPCR (p<0.001), showing a significant reduction in Nrf2 expression. Notably, this knockdown resulted in increased expression of BAT markers, including PGC-1α (p = 0.012), Dio2 (p = 0.020), and PRDM16 (p = 0.001), at both mRNA (PGC-1α [p = 0.012], Dio2 [p = 0.020], and PRDM16 [p = 0.001]) and protein (PGC-1α [p = 0.001]; Dio2 [p = 0.003]; PRDM16 [p = 0.007])levels. Conversely, WAT markers such as BMP4 (mRNA: p = 0.01; WB: p = 0.001), resistin (mRNA: p = 0.016; WB: p = 0.004), and Rb1 (mRNA: p = 0.03; WB: p = 0.003) were significantly downregulated. Additionally, levels of Cycs (mRNA: p = 0.024; WB: p = 0.037) and UCP1 (mRNA: p = 0.024; WB: p = 0.023) were elevated, indicating enhanced mitochondrial function and metabolic activity (P < 0.05). The knockdown also affected longevity-related proteins, Sirt1 (WB: p = 0.018) and AMPKα (WB: p = 0.021), underscoring Nrf2's role in metabolic regulation. CONCLUSION Nrf2 knockdown in 3T3-L1 adipocytes promotes a transition towards a brown adipose phenotype and enhances the expression of longevity-related factors, suggesting Nrf2 as a potential therapeutic target for addressing aging-related metabolic decline.
Collapse
Affiliation(s)
- Sai Zhang
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050030, China
| | - Bing Han
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050030, China
| | - Xue Wang
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050030, China
| | - Xiaoyang Yuan
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050030, China.
| |
Collapse
|
4
|
Yeo RX, Mau T, Ross ZM, Edenhoffer NP, Liu J, Barnes HN, Lui LY, Adkins JN, Sanford JA, Seldin MM, Viesi CH, Zhou M, Gregory HL, Toledo FGS, Stefanovic-Racic M, Lyles M, Wood AN, Mattila PE, Blakley EA, Miljkovic I, Cawthon PM, Newman AB, Kritchevsky SB, Cummings SR, Goodpaster BH, Justice JN, Kershaw EE, Sparks LM. Investigating the role of adipose tissue in mobility and aging: design and methods of the Adipose Tissue ancillary to the Study of Muscle, Mobility, and Aging (SOMMA-AT). J Gerontol A Biol Sci Med Sci 2025; 80:glaf015. [PMID: 39886989 DOI: 10.1093/gerona/glaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Age-related changes in adipose tissue affect chronic medical diseases and mobility disability but mechanism remains poorly understood. The goal of this study is to define methods for phenotyping unique characteristics of adipose tissue from older adults. METHODS Older adults enrolled in study of muscle, mobility, and aging selected for the adipose tissue ancillary (SOMMA-AT; N = 210, 52.38% women, 76.12 ± 4.37 years) were assessed for regional adiposity by whole-body magnetic resonance (AMRA) and underwent a needle-aspiration biopsy of abdominal subcutaneous adipose tissue (ASAT). ASAT biopsies were flash frozen, fixed, or processed for downstream applications and deposited at the biorepository. Biopsy yields, qualitative features, adipocyte sizes, and concentration of adipokines secreted in ASAT explant conditioned media were measured. Inter-measure Spearman correlations were determined. RESULTS Regional, but not total, adiposity differed by sex: women had greater ASAT mass (8.20 ± 2.73 kg, p < .001) and biopsy yield (3.44 ± 1.81 g, p < .001) than men (ASAT = 5.95 ± 2.30 kg, biopsy = 2.30 ± 1.40 g). ASAT mass correlated with leptin (r = 0.54, p < .001) and not resistin (p = .248) and adiponectin (p = .353). Adipocyte area correlated with ASAT mass (r = 0.34, p < .001), BMI (r = 0.33, p < .001), adiponectin (r = -0.22, p = .005) and leptin (r = 0.18, p = .024) but not with resistin (p = .490). CONCLUSION In addition to the detailed ASAT biopsy processing in this report, we found that adipocyte area correlated with ASAT mass, and both measures related to some key adipokines in the explant conditioned media. These results, methods, and biological repositories underscore the potential of this unique cohort to impact the understanding of aging adipose biology on disease, disability, and other aging tissues.
Collapse
Affiliation(s)
- Reichelle X Yeo
- AdventHealth Translational Research Institute, Orlando, Florida, USA
| | - Theresa Mau
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
- Department of Epidemiology, San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Zana M Ross
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nicholas P Edenhoffer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jingfang Liu
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Haley N Barnes
- Department of Epidemiology, San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Li-Yung Lui
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
- Department of Epidemiology, San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Joshua N Adkins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - James A Sanford
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, California, USA
| | - Carlos H Viesi
- Department of Biological Chemistry, University of California, Irvine, Irvine, California, USA
| | - Mingqi Zhou
- Department of Biological Chemistry, University of California, Irvine, Irvine, California, USA
| | - Heather L Gregory
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Frederico G S Toledo
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Maja Stefanovic-Racic
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mary Lyles
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ashlee N Wood
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Polly E Mattila
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Iva Miljkovic
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Peggy M Cawthon
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
- Department of Epidemiology, San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Anne B Newman
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Steven R Cummings
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
- Department of Epidemiology, San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Bret H Goodpaster
- AdventHealth Translational Research Institute, Orlando, Florida, USA
| | - Jamie N Justice
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- XPRIZE Foundation, Culver City, California, USA
| | - Erin E Kershaw
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lauren M Sparks
- AdventHealth Translational Research Institute, Orlando, Florida, USA
| |
Collapse
|
5
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
6
|
Jeon YG, Kim JB. The cellular basis for middle-age spread. Science 2025; 388:360. [PMID: 40273264 DOI: 10.1126/science.adx1198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Age-specific adipocyte progenitors drive visceral adipose tissue expansion in middle age.
Collapse
Affiliation(s)
- Yong Geun Jeon
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- National Leader Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
7
|
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
|
8
|
Li J, Bea JW, LaMonte M, Jiang L, Reding K, Garcia L, Manson JAE, Follis S, Odegaard AO. Effects of moderate/vigorous activity on 3-year body composition changes in postmenopausal women: a target trial emulation. Am J Epidemiol 2025; 194:1032-1042. [PMID: 38992341 DOI: 10.1093/aje/kwae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 06/05/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Postmenopausal women experience significant changes in body composition, particularly abdominal adipose tissue (AAT) deposition patterns, which influence cardiometabolic risk. Physical activity has demonstrable effects on body composition and overall health; however, there is little evidence for how physical activity influences AAT patterns and body composition in postmenopausal women. We emulated a target trial of physical activity interventions, including the 2018 Physical Activity Guidelines for Americans recommendations (≥150 minutes/week), on 3-year changes in AAT and body composition. We analyzed data from 4451 postmenopausal women aged 50-79 years in the Women's Health Initiative (WHI) with repeated whole body Dual X-Ray Absorptiometry (DXA) scans with derived abdominal visceral (VAT) and subcutaneous adipose tissue (SAT). The mean AAT and body composition measures were estimated with the parametric-g formula. Over 3 years, interventions of increasing minutes of moderate activity would result in dose-dependent reductions in AAT, overall body fat and increases in lean soft tissue, with the greatest estimated benefit at the 2018 physical activity guideline recommendations. Compared to no intervention, if all participants had adhered to ≥150 mins/week of moderate physical activity, they would have 16.8 cm2 lower VAT (95% CI: -23.1, -10.4), 26.8 cm2 lower SAT (95% CI: -36.3, -17.3), 1.3% lower total body fat (95% CI: -1.8, -0.7), 1.2% higher total lean soft tissue (95% CI: 0.7-1.8), and 2.6 kg lower bodyweight (95% CI, -3.6, -1.5). We saw similar patterns in vigorous-intensity activity interventions. These results suggest that postmenopausal women who adhere to physical activity guideline recommendations would experience beneficial body composition changes over 3 years.
Collapse
Affiliation(s)
- Jiarui Li
- Epidemiology and Biostatistics, University of California Irvine, Irvine, CA, United States
| | - Jennifer W Bea
- Department of Health Promotion Sciences, University of Arizona Cancer Center, Tucson, AZ, United States
| | - Michael LaMonte
- Department of Epidemiology and Environmental Health, Social and Preventive Medicine, University of Buffalo, Buffalo, NY, United States
| | - Luohua Jiang
- Epidemiology and Biostatistics, University of California Irvine, Irvine, CA, United States
| | - Kerryn Reding
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Lorena Garcia
- Department of Public Health Sciences, University of California, Davis, CA, United States
| | - Jo Ann E Manson
- Division of Preventive Medicine, Brigham and Women's Hospital, Professor of Medicine and the Michael and Lee Bell Professor of Women's Health, Harvard Medical School Boston, Department of Epidemiology, Harvard University, MA, United States
| | - Shawna Follis
- Department of Medicine, Stanford University, Palo Alto, CA, United States
| | - Andrew O Odegaard
- Epidemiology and Biostatistics, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
9
|
Wang S, Huo T, Lu M, Zhao Y, Zhang J, He W, Chen H. Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies. Cells 2025; 14:499. [PMID: 40214453 PMCID: PMC11987807 DOI: 10.3390/cells14070499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Cellular senescence is an irreversible state of cell cycle arrest. Senescent cells (SCs) accumulate in the body with age and secrete harmful substances known as the senescence-associated secretory phenotype (SASP), causing chronic inflammation; at the same time, chronic inflammation leads to a decrease in immune system function, known as immunosenescence, which further accelerates the aging process. Cellular senescence and immunosenescence are closely related to a variety of chronic diseases, including cardiovascular diseases, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Studying the mechanisms of cellular senescence and immunosenescence and developing targeted interventions are crucial for improving the immune function and quality of life of elderly people. Here, we review a series of recent studies focusing on the molecular mechanisms of cellular senescence and immunosenescence, the regulation of aging by the immune system, and the latest advances in basic and clinical research on senolytics. We summarize the cellular and animal models related to aging research, as well as the mechanisms, strategies, and future directions of aging interventions from an immunological perspective, with the hope of laying the foundation for developing novel and practical anti-aging therapies.
Collapse
Affiliation(s)
- Shuaiqi Wang
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Tong Huo
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Mingyang Lu
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Yueqi Zhao
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Jianmin Zhang
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
| | - Wei He
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Hui Chen
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
| |
Collapse
|
10
|
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
|
11
|
Loh NY, Vasan SK, Rosoff DB, Roberts E, van Dam AD, Verma M, Phillips D, Wesolowska-Andersen A, Neville MJ, Noordam R, Ray DW, Tobias JH, Gregson CL, Karpe F, Christodoulides C. LRP5 promotes adipose progenitor cell fitness and adipocyte insulin sensitivity. COMMUNICATIONS MEDICINE 2025; 5:51. [PMID: 40000740 PMCID: PMC11862225 DOI: 10.1038/s43856-025-00774-1] [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: 03/04/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND WNT signaling plays a key role in postnatal bone formation. Individuals with gain-of-function mutations in the WNT co-receptor LRP5 exhibit increased lower-body fat mass and potentially enhanced glucose metabolism, alongside high bone mass. However, the mechanisms by which LRP5 regulates fat distribution and its effects on systemic metabolism remain unclear. This study aims to explore the role of LRP5 in adipose tissue biology and its impact on metabolism. METHODS Metabolic assessments and imaging were conducted on individuals with gain- and loss-of-function LRP5 mutations, along with age- and BMI-matched controls. Mendelian randomization analyses were used to investigate the relationship between bone, fat distribution, and systemic metabolism. Functional studies and RNA sequencing were performed on abdominal and gluteal adipose cells with LRP5 knockdown. RESULTS Here we show that LRP5 promotes lower-body fat distribution and enhances systemic and adipocyte insulin sensitivity through cell-autonomous mechanisms, independent of its bone-related functions. LRP5 supports adipose progenitor cell function by activating WNT/β-catenin signaling and preserving valosin-containing protein (VCP)-mediated proteostasis. LRP5 expression in adipose progenitors declines with age, but gain-of-function LRP5 variants protect against age-related fat loss in the lower body. CONCLUSIONS Our findings underscore the critical role of LRP5 in regulating lower-body fat distribution and insulin sensitivity, independent of its effects on bone. Pharmacological activation of LRP5 in adipose tissue may offer a promising strategy to prevent age-related fat redistribution and metabolic disorders.
Collapse
Affiliation(s)
- Nellie Y Loh
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Senthil K Vasan
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel B Rosoff
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Emile Roberts
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Andrea D van Dam
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Manu Verma
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel Phillips
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Agata Wesolowska-Andersen
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Matt J Neville
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - David W Ray
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Celia L Gregson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Fredrik Karpe
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Constantinos Christodoulides
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK.
| |
Collapse
|
12
|
Natarajan D, Ekambaram S, Tarantini S, Nagaraja RY, Yabluchanskiy A, Hedrick AF, Awasthi V, Subramanian M, Csiszar A, Balasubramanian P. Chronic β3 adrenergic agonist treatment improves neurovascular coupling responses, attenuates blood-brain barrier leakage and neuroinflammation, and enhances cognition in aged mice. Aging (Albany NY) 2025; 17:448-463. [PMID: 39976587 PMCID: PMC11892913 DOI: 10.18632/aging.206203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025]
Abstract
Microvascular endothelial dysfunction, characterized by impaired neurovascular coupling, reduced glucose uptake, blood-brain barrier disruption, and microvascular rarefaction, plays a critical role in the pathogenesis of age-related vascular cognitive impairment (VCI). Emerging evidence points to non-cell autonomous mechanisms mediated by adverse circulating milieu (an increased ratio of pro-geronic to anti-geronic circulating factors) in the pathogenesis of endothelial dysfunction leading to impaired cerebral blood flow and cognitive decline in the aging population. In particular, age-related adipose dysfunction contributes, at least in part, to an unfavorable systemic milieu characterized by chronic hyperglycemia, hyperinsulinemia, dyslipidemia, and altered adipokine profile, which together contribute to microvascular endothelial dysfunction. Hence, in the present study, we aimed to test whether thermogenic stimulation, an intervention known to improve adipose and systemic metabolism by increasing cellular energy expenditure, could mitigate brain endothelial dysfunction and improve cognition in the aging population. Eighteen-month-old C57BL/6J mice were treated with saline or β3-adrenergic agonist (CL 316, 243, CL) for 6 weeks followed by functional analysis to assess endothelial function and cognition. CL treatment improved neurovascular coupling responses and rescued brain glucose uptake in aged animals. In addition, CL treatment also attenuated blood-brain barrier leakage and associated neuroinflammation in the cortex and increased microvascular density in the hippocampus of aged mice. More importantly, these beneficial changes in microvascular function translated to improved cognitive performance in aged mice. Our results suggest that β3-adrenergic agonist treatment improves multiple aspects of cerebromicrovascular function and can be potentially repurposed for treating age-associated cognitive decline.
Collapse
Affiliation(s)
- Duraipandy Natarajan
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, 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
| | - Shoba Ekambaram
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Stefano Tarantini
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Raghavendra Y. Nagaraja
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, 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
| | - Andriy Yabluchanskiy
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Andria F. Hedrick
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Madhan Subramanian
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 73104, USA
| | - Anna Csiszar
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, 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
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Priya Balasubramanian
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, 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
| |
Collapse
|
13
|
Fu X, Zhao Y, Cui X, Huang S, Lv Y, Li C, Gong F, Yang Z, Yang X, Xiao R. Cxcl9 modulates aging associated microvascular metabolic and angiogenic dysfunctions in subcutaneous adipose tissue. Angiogenesis 2025; 28:17. [PMID: 39934436 PMCID: PMC11813824 DOI: 10.1007/s10456-025-09970-y] [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: 10/29/2024] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
Microvascular aging, predominantly driven by endothelial cells (ECs) dysfunction, is a critical early event in cardiovascular diseases. However, the specific effects of aging on ECs across the microvascular network segments and the associated mechanisms are not fully understood. In this study, we detected a microvascular rarefaction and a decreased proportion of venular ECs in the subcutaneous adipose tissue of aged mice using light-sheet immunofluorescence microscopy and single-cell RNA sequencing. Moreover, aged ECs, especially in the venular subtype, exhibited a pseudotemporal transition to a terminal state characterized by diminished oxidative phosphorylation and strengthened cytokine signaling. Metabolic flux balance analysis predicted that among the 13 differentially expressed cytokines identified in aged EC subpopulations, Cxcl9 was strongly correlated with impaired oxidative phosphorylation in aged ECs. It was further validated using microvascular ECs treated with Cxcl9. Notably, the G protein-coupled receptor signaling pathway was subsequently suppressed, in which Aplnr suppression was also observed in aged ECs, contributing to their impaired energy metabolism and reduced angiogenesis. Based on these findings, we propose Cxcl9 as a biomarker for aging-related dysfunction of microvascular ECs, suggesting that targeting Cxcl9 signaling may help combat microvascular aging.
Collapse
Affiliation(s)
- Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Yu Zhao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Xiwei Cui
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Siyuan Huang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China
| | - Yanze Lv
- Department of Hemangioma and Vascular Malformation of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
| | - Chen Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Fuxing Gong
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Xiaonan Yang
- Department of Hemangioma and Vascular Malformation of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China.
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China.
| |
Collapse
|
14
|
de Oliveira Silva T, Lunardon G, Lino CA, de Almeida Silva A, Zhang S, Irigoyen MCC, Lu YW, Mably JD, Barreto-Chaves MLM, Wang DZ, Diniz GP. Senescent cell depletion alleviates obesity-related metabolic and cardiac disorders. Mol Metab 2025; 91:102065. [PMID: 39557194 PMCID: PMC11636344 DOI: 10.1016/j.molmet.2024.102065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/06/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Obesity is a major contributor to metabolic and cardiovascular disease. Although senescent cells have been shown to accumulate in adipose tissue, the role of senescence in obesity-induced metabolic disorders and in cardiac dysfunction is not yet clear; therefore, the therapeutic potential of managing senescence in obesity-related metabolic and cardiac disorders remains to be fully defined. OBJECTIVE We investigated the beneficial effects of a senolytic cocktail (dasatinib and quercetin) on senescence and its influence on obesity-related parameters. METHODS AND RESULTS We found that the increase in body weight and adiposity, glucose intolerance, insulin resistance, dyslipidemia, hyperleptinemia, and hepatic disorders which were induced by an obesogenic diet were alleviated by senolytic cocktail treatment in mice. Treatment with senolytic compounds eliminated senescent cells, counteracting the activation of the senescence program and DNA damage in white adipose tissue (WAT) observed with an obesogenic diet. Moreover, the senolytic cocktail prevented the brown adipose tissue (BAT) whitening and increased the expression of the thermogenic gene profile in BAT and pWAT. In the hearts of obese mice, senolytic combination abolished myocardial maladaptation, reducing the senescence-associated secretory phenotype (SASP) and DNA damage, repressing cardiac hypertrophy, and improving diastolic dysfunction. Additionally, we showed that treatment with the senolytic cocktail corrected gene expression programs associated with fatty acid metabolism, oxidative phosphorylation, the P53 pathway, and DNA repair, which were all downregulated in obese mice. CONCLUSIONS Collectively, these data suggest that a senolytic cocktail can prevent the activation of the senescence program in the heart and WAT and activate the thermogenic program in BAT. Our results suggest that targeting senescent cells may be a novel therapeutic strategy for alleviating obesity-related metabolic and cardiac disorders.
Collapse
Affiliation(s)
- Tábatha de Oliveira Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Guilherme Lunardon
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Caroline A Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Amanda de Almeida Silva
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Shiju Zhang
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | | | - Yao Wei Lu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John D Mably
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | | | - Da-Zhi Wang
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Gabriela P Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
15
|
Pangrazzi L, Meryk A. Molecular and Cellular Mechanisms of Immunosenescence: Modulation Through Interventions and Lifestyle Changes. BIOLOGY 2024; 14:17. [PMID: 39857248 PMCID: PMC11760833 DOI: 10.3390/biology14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Immunosenescence, the age-related decline in immune function, is a complex biological process with profound implications for health and longevity. This phenomenon, characterized by alterations in both innate and adaptive immunity, increases susceptibility to infections, reduces vaccine efficacy, and contributes to the development of age-related diseases. At the cellular level, immunosenescence manifests as decreased production of naive T and B cells, accumulation of memory and senescent cells, thymic involution, and dysregulated cytokine production. Recent advances in molecular biology have shed light on the underlying mechanisms of immunosenescence, including telomere attrition, epigenetic alterations, mitochondrial dysfunction, and changes in key signaling pathways such as NF-κB and mTOR. These molecular changes lead to functional impairments in various immune cell types, altering their proliferative capacity, differentiation, and effector functions. Emerging research suggests that lifestyle factors may modulate the rate and extent of immunosenescence at both cellular and molecular levels. Physical activity, nutrition, stress management, and sleep patterns have been shown to influence immune cell function, inflammatory markers, and oxidative stress in older adults. This review provides a comprehensive analysis of the molecular and cellular mechanisms underlying immunosenescence and explores how lifestyle interventions may impact these processes. We will examine the current understanding of immunosenescence at the genomic, epigenomic, and proteomic levels, and discuss how various lifestyle factors can potentially mitigate or partially reverse aspects of immune aging. By integrating recent findings from immunology, gerontology, and molecular biology, we aim to elucidate the intricate interplay between lifestyle and immune aging at the molecular level, potentially informing future strategies for maintaining immune competence in aging populations.
Collapse
Affiliation(s)
- Luca Pangrazzi
- Institute for Biomedical Aging Research, Faculty of Biology, University of Innsbruck, 6020 Innsbruck, Austria;
| | - Andreas Meryk
- Department of Pediatrics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
16
|
Huang B, Gao Y, Wu L. Assessment of body composition and prediction of infectious pancreatic necrosis via non-contrast CT radiomics and deep learning. Front Microbiol 2024; 15:1509915. [PMID: 39735191 PMCID: PMC11671486 DOI: 10.3389/fmicb.2024.1509915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Aim The current study aims to delineate subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), the sacrospinalis muscle, and all abdominal musculature at the L3-L5 vertebral level from non-contrast computed tomography (CT) imagery using deep learning algorithms. Subsequently, radiomic features are collected from these segmented images and subjected to medical interpretation. Materials and methods This retrospective analysis includes a cohort of 315 patients diagnosed with acute necrotizing pancreatitis (ANP) who had undergone comprehensive whole-abdomen CT scans. The no new net (nnU-Net) architecture was adopted for the imagery segmentation, while Python scripts were employed to derive radiomic features from the segmented non-contrast CT images. In light of the intrinsic medical relevance of specific features, two categories were selected for analysis: first-order statistics and morphological characteristics. A correlation analysis was conducted, and statistically significant features were subjected to medical scrutiny. Results With respect to VAT, skewness (p = 0.004) and uniformity (p = 0.036) emerged as statistically significant; for SAT, significant features included skewness (p = 0.023), maximum two-dimensional (2D) diameter slice (p = 0.020), and maximum three-dimensional (3D) diameter (p = 0.044); for the abdominal muscles, statistically significant metrics were the interquartile range (IQR; p = 0.023), mean absolute deviation (p = 0.039), robust mean absolute deviation (p = 0.015), elongation (p = 0.025), sphericity (p = 0.010), and surface volume ratio (p = 0.014); and for the sacrospinalis muscle, significant indices comprised the IQR (p = 0.018), mean absolute deviation (p = 0.049), robust mean absolute deviation (p = 0.025), skewness (p = 0.008), maximum 2D diameter slice (p = 0.008), maximum 3D diameter (p = 0.005), sphericity (p = 0.011), and surface volume ratio (p = 0.005). Conclusion Diminished localized deposition of VAT and SAT, homogeneity in the VAT and SAT density, augmented SAT volume, and a dispersed and heterogeneous distribution of abdominal muscle density are identified as risk factors for infectious pancreatic necrosis (IPN).
Collapse
Affiliation(s)
- Bingyao Huang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi Gao
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
Lv D, Ren Y, Chen J, Pang Z, Tang Y, Zhang L, Yan L, Ai X, Xv X, Wang D, Cai Z. Protective Effects of Exogenous Melatonin Administration on White Fat Metabolism Disruption Induced by Aging and a High-Fat Diet in Mice. Antioxidants (Basel) 2024; 13:1500. [PMID: 39765828 PMCID: PMC11672923 DOI: 10.3390/antiox13121500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Obesity has emerged as a major risk factor for human health, exacerbated by aging and changes in dietary habits. It represents a significant health challenge, particularly for older people. While numerous studies have examined the effects of obesity and aging on fat metabolism independently, research on their combined effects is limited. In the present study, the protective action against white fat accumulation after a high-fat diet (HFD) exerted by exogenous melatonin, a circadian hormone endowed with antioxidant properties also involved in fat metabolism, was investigated in a mouse model. For this purpose, a battery of tests was applied before and after the dietary and melatonin treatments of the animals, including epididymal white adipose tissue (eWAT) histological evaluations, transcriptomic and lipidomic analyses, real-time PCR tests, immunofluorescence staining, Western blot, the appraisal of serum melatonin levels, and transmission electron microscopy. This study found that aged mice on a high-fat diet (HFD) showed increased lipid deposition, inflammation, and reduced antioxidant glutathione (GSH) levels compared to younger mice. Lipidomic and transcriptomic analyses revealed elevated triglycerides, diglycerides, ceramides, and cholesterol, along with decreased sphingomyelin and fatty acids in eWAT. The genes linked to inflammation, NF-κB signaling, autophagy, and lipid metabolism, particularly the melatonin and glutathione pathways, were significantly altered. The aged HFD mice also exhibited reduced melatonin levels in serum and eWAT. Melatonin supplementation reduced lipid deposition, increased melatonin and GSH levels, and upregulated AANAT and MTNR1A expression in eWAT, suggesting that melatonin alleviates eWAT damage via the MTNR1A pathway. It also suppressed inflammatory markers (e.g., TNF-α, NLRP3, NF-κB, IL-1β, and CEBPB) and preserved mitochondrial function through enhanced mitophagy. This study highlights how aging and HFD affect lipid metabolism and gene expression, offering potential intervention strategies. These findings provide important insights into the mechanisms of fat deposition associated with aging and a high-fat diet, suggesting potential intervention strategies.
Collapse
Affiliation(s)
- Dongying Lv
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (D.L.); (Y.T.); (L.Z.); (X.A.); (X.X.)
| | - Yujie Ren
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.R.); (J.C.); (Z.P.)
| | - Jiayan Chen
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.R.); (J.C.); (Z.P.)
| | - Ziyao Pang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.R.); (J.C.); (Z.P.)
| | - Yaxuan Tang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (D.L.); (Y.T.); (L.Z.); (X.A.); (X.X.)
| | - Lizong Zhang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (D.L.); (Y.T.); (L.Z.); (X.A.); (X.X.)
| | - Laiqing Yan
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Xiufeng Ai
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (D.L.); (Y.T.); (L.Z.); (X.A.); (X.X.)
| | - Xiaoping Xv
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (D.L.); (Y.T.); (L.Z.); (X.A.); (X.X.)
| | - Dejun Wang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (D.L.); (Y.T.); (L.Z.); (X.A.); (X.X.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.R.); (J.C.); (Z.P.)
| | - Zhaowei Cai
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (D.L.); (Y.T.); (L.Z.); (X.A.); (X.X.)
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.R.); (J.C.); (Z.P.)
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
18
|
Xu W, Chen H, Xiao H. mTORC2: A neglected player in aging regulation. J Cell Physiol 2024; 239:e31363. [PMID: 38982866 DOI: 10.1002/jcp.31363] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/21/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024]
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that plays a pivotal role in various biological processes, through integrating external and internal signals, facilitating gene transcription and protein translation, as well as by regulating mitochondria and autophagy functions. mTOR kinase operates within two distinct protein complexes known as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which engage separate downstream signaling pathways impacting diverse cellular processes. Although mTORC1 has been extensively studied as a pro-proliferative factor and a pro-aging hub if activated aberrantly, mTORC2 received less attention, particularly regarding its implication in aging regulation. However, recent studies brought increasing evidence or clues for us, which implies the associations of mTORC2 with aging, as the genetic elimination of unique subunits of mTORC2, such as RICTOR, has been shown to alleviate aging progression in comparison to mTORC1 inhibition. In this review, we first summarized the basic characteristics of mTORC2, including its protein architecture and signaling network. We then focused on reviewing the molecular signaling regulation of mTORC2 in cellular senescence and organismal aging, and proposed the multifaceted regulatory characteristics under senescent and nonsenescent contexts. Next, we outlined the research progress of mTOR inhibitors in the field of antiaging and discussed future prospects and challenges. It is our pleasure if this review article could provide meaningful information for our readers and call forth more investigations working on this topic.
Collapse
Affiliation(s)
- Weitong Xu
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Honghan Chen
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Whytock KL, Divoux A, Sun Y, Pino MF, Yu G, Jin CA, Robino JJ, Plekhanov A, Varlamov O, Smith SR, Walsh MJ, Sparks LM. Aging human abdominal subcutaneous white adipose tissue at single cell resolution. Aging Cell 2024; 23:e14287. [PMID: 39141531 PMCID: PMC11561672 DOI: 10.1111/acel.14287] [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: 02/06/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
White adipose tissue (WAT) is a robust energy storage and endocrine organ critical for maintaining metabolic health as we age. Our aim was to identify cell-specific transcriptional aberrations that occur in WAT with aging. We leveraged full-length snRNA-Seq and histology to characterize the cellular landscape of human abdominal subcutaneous WAT in a prospective cohort of 10 younger (≤30 years) and 10 older individuals (≥65 years) balanced for sex and body mass index (BMI). The older group had greater cholesterol, very-low-density lipoprotein, triglycerides, thyroid stimulating hormone, and aspartate transaminase compared to the younger group (p < 0.05). We highlight that aging WAT is associated with adipocyte hypertrophy, increased proportions of lipid-associated macrophages and mast cells, an upregulation of immune responses linked to fibrosis in pre-adipocyte, adipocyte, and vascular populations, and highlight CXCL14 as a biomarker of these processes. We show that older WAT has elevated levels of senescence marker p16 in adipocytes and identify the adipocyte subpopulation driving this senescence profile. We confirm that these transcriptional and phenotypical changes occur without overt fibrosis and in older individuals that have comparable WAT insulin sensitivity to the younger individuals.
Collapse
Affiliation(s)
- K. L. Whytock
- Translational Research Institute, AdventHealthOrlandoFloridaUSA
| | - A. Divoux
- Translational Research Institute, AdventHealthOrlandoFloridaUSA
| | - Y. Sun
- Icahn School of Medicine at Mount SinaiNew York CityNew YorkUSA
| | - M. F. Pino
- Translational Research Institute, AdventHealthOrlandoFloridaUSA
| | - G. Yu
- Translational Research Institute, AdventHealthOrlandoFloridaUSA
| | - C. A. Jin
- Department of Genetics, School of MedicineStanford UniversityStanfordCaliforniaUSA
| | - J. J. Robino
- Divisions of Metabolic Health and DiseaseOregon National Primate Research CenterBeavertonOregonUSA
| | - A. Plekhanov
- Divisions of Metabolic Health and DiseaseOregon National Primate Research CenterBeavertonOregonUSA
| | - O. Varlamov
- Divisions of Metabolic Health and DiseaseOregon National Primate Research CenterBeavertonOregonUSA
| | - S. R. Smith
- Translational Research Institute, AdventHealthOrlandoFloridaUSA
| | - M. J. Walsh
- Icahn School of Medicine at Mount SinaiNew York CityNew YorkUSA
| | - L. M. Sparks
- Translational Research Institute, AdventHealthOrlandoFloridaUSA
| |
Collapse
|
20
|
Dorf N, Maciejczyk M. Skin senescence-from basic research to clinical practice. Front Med (Lausanne) 2024; 11:1484345. [PMID: 39493718 PMCID: PMC11527680 DOI: 10.3389/fmed.2024.1484345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
The most recognizable implications of tissue aging manifest themselves on the skin. Skin laxity, roughness, pigmentation disorders, age spots, wrinkles, telangiectasia or hair graying are symptoms of physiological aging. Development of the senescent phenotype depends on the interaction between aging cells and remodeling of the skin's extracellular matrix (ECM) that contains collagen and elastic fiber. Aging changes occur due to the combination of both endogenous (gene mutation, cellular metabolism or hormonal agents) and exogenous factors (ultraviolet light, environmental pollutants, and unsuitable diet). However, overproduction of mitochondrial reactive oxygen species (ROS) is a key factor driving cellular senescence. Aging theories have disclosed a range of diverse molecular mechanisms that are associated with cellular senescence of the body. Theories best supported by evidence include protein glycation, oxidative stress, telomere shortening, cell cycle arrest, and a limited number of cell divisions. Accumulation of the ECM damage is suggested to be a key factor in skin aging. Every cell indicates a functional and morphological change that may be used as a biomarker of senescence. Senescence-associated β-galactosidase (SA-β-gal), cell cycle inhibitors (p16INK4a, p21CIP1, p27, p53), DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS), senescence-associated heterochromatin foci (SAHF), shortening of telomeres or downregulation of lamina B1 constitute just an example of aging biomarkers known so far. Aging may also be assessed non-invasively through measuring the skin fluorescence of advanced glycation end-products (AGEs). This review summarizes the recent knowledge on the pathogenesis and clinical conditions of skin aging as well as biomarkers of skin senescence.
Collapse
Affiliation(s)
- Natalia Dorf
- Independent Laboratory of Cosmetology, Medical University of Białystok, Bialystok, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Białystok, Bialystok, Poland
| |
Collapse
|
21
|
Pellegrini V, La Grotta R, Carreras F, Giuliani A, Sabbatinelli J, Olivieri F, Berra CC, Ceriello A, Prattichizzo F. Inflammatory Trajectory of Type 2 Diabetes: Novel Opportunities for Early and Late Treatment. Cells 2024; 13:1662. [PMID: 39404426 PMCID: PMC11476093 DOI: 10.3390/cells13191662] [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] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Low-grade inflammation (LGI) represents a key driver of type 2 diabetes (T2D) and its associated cardiovascular diseases (CVDs). Indeed, inflammatory markers such as hs-CRP and IL-6 predict the development of T2D and its complications, suggesting that LGI already increases before T2D diagnosis and remains elevated even after treatment. Overnutrition, unhealthy diets, physical inactivity, obesity, and aging are all recognized triggers of LGI, promoting insulin resistance and sustaining the pathogenesis of T2D. Once developed, and even before frank appearance, people with T2D undergo a pathological metabolic remodeling, with an alteration of multiple CVD risk factors, i.e., glycemia, lipids, blood pressure, and renal function. In turn, such variables foster a range of inflammatory pathways and mechanisms, e.g., immune cell stimulation, the accrual of senescent cells, long-lasting epigenetic changes, and trained immunity, which are held to chronically fuel LGI at the systemic and tissue levels. Targeting of CVD risk factors partially ameliorates LGI. However, some long-lasting inflammatory pathways are unaffected by common therapies, and LGI burden is still increased in many T2D patients, a phenomenon possibly underlying the residual inflammatory risk (i.e., having hs-CRP > 2 mg/dL despite optimal LDL cholesterol control). On the other hand, selected disease-modifying drugs, e.g., GLP-1RA, seem to also act on the pathogenesis of T2D, curbing the inflammatory trajectory of the disease and possibly preventing it if introduced early. In addition, selected trials demonstrated the potential of canonical anti-inflammatory therapies in reducing the rate of CVDs in patients with this condition or at high risk for it, many of whom had T2D. Since colchicine, an inhibitor of immune cell activation, is now approved for the prevention of CVDs, it might be worth exploring a possible therapeutic paradigm to identify subjects with T2D and an increased LGI burden to treat them with this drug. Upcoming studies will reveal whether disease-modifying drugs reverse early T2D by suppressing sources of LGI and whether colchicine has a broad benefit in people with this condition.
Collapse
Affiliation(s)
- Valeria Pellegrini
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | - Rosalba La Grotta
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | - Francesca Carreras
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | - Angelica Giuliani
- Cardiac Rehabilitation Unit of Bari Institute, Istituti Clinici Scientifici Maugeri IRCCS, 70124 Bari, Italy
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60127 Ancona, Italy; (J.S.); (F.O.)
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60127 Ancona, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60127 Ancona, Italy; (J.S.); (F.O.)
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60127 Ancona, Italy
| | | | - Antonio Ceriello
- IRCCS MultiMedica, Via Fantoli 16/15, 20138 Milan, Italy; (V.P.); (R.L.G.)
| | | |
Collapse
|
22
|
Li C, Zhang Y, Wang Y, Gu C, Li B, Ma M, Xu X, Chen Y, Zheng Z. Imaging-based body fat distribution and diabetic retinopathy in general US population with diabetes: an NHANES analysis (2003-2006 and 2011-2018). Nutr Diabetes 2024; 14:53. [PMID: 39004614 PMCID: PMC11247072 DOI: 10.1038/s41387-024-00308-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Limited studies have investigated the correlation between fat distribution and the risk of diabetic retinopathy (DR) in the general population with diabetes. The relationship between obesity and DR remains inconclusive, possibly due to using simple anthropometric measures to define obesity. This study investigates the relationships between the android-to-gynoid fat ratio (A/G ratio, measured using dual-energy X-ray absorptiometry) and DR within the US population with diabetes. METHODS The study used a population-based, cross-sectional approach based on the 2003-2006 and 2011-2018 data of the National Health and Nutrition Examination Survey (NHANES). Multivariable logistic regression analyses were performed on participants with diabetes to evaluate the contribution of body mass index (BMI), waist-to-height ratio (WHtR), and A/G ratio to the prevalence of DR. RESULTS The prevalence of DR was 22.2, 21.2, and 17.6% among participants with A/G ratios <1.0, 1.0-1.2, and ≥1.2, respectively. After adjusting sex, age, ethnicity, diabetes duration, hemoglobin A1c level, blood pressure level, and non-high-density lipoprotein cholesterol level, a higher A/G ratio (≥1.2) was independently associated with decreased odds of DR (odds ratio [OR], 0.565; 95% CI: 0.372-0.858) compared with the A/G ratio of 1.0-1.2. Associations between a higher A/G ratio and DR remained statistically significant after adjusting for BMI (OR, 0.567; 95% CI: 0.373-0.861) and WHtR (OR, 0.586; 95% CI: 0.379-0.907). Moreover, these associations remained statistically significant in analyses using the ethnic-specific tertiles for the A/G ratio. In sex-stratified models, these correlations remained in males. There was a significant inverse association between the A/G ratio and diabetes duration in males, which persisted after multivariable adjustments (p < 0.05). CONCLUSIONS A novel finding indicates that a higher A/G ratio is associated with a reduced likelihood of DR in males with diabetes. The results from NHANES underscore the importance of considering imaging-based fat distribution as a critical indicator in clinical practice.
Collapse
Affiliation(s)
- Chenxin Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Clinical Research Center for Eye Diseases, Shanghai Key Clinical Specialty, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Yili Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, 200080, China
| | - Yujie Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Clinical Research Center for Eye Diseases, Shanghai Key Clinical Specialty, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Chufeng Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Clinical Research Center for Eye Diseases, Shanghai Key Clinical Specialty, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Bo Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Clinical Research Center for Eye Diseases, Shanghai Key Clinical Specialty, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Mingming Ma
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Clinical Research Center for Eye Diseases, Shanghai Key Clinical Specialty, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Xiaoyin Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Clinical Research Center for Eye Diseases, Shanghai Key Clinical Specialty, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| | - Yongdong Chen
- Department of Ophthalmology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Clinical Research Center for Eye Diseases, Shanghai Key Clinical Specialty, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
- Ningde Municipal Hospital, Ningde Normal University, Ningde, China.
- Fujian Medical University, Fuzhou, China.
| |
Collapse
|
23
|
Natarajan D, Ekambaram S, Tarantini S, Yelahanka Nagaraja R, Yabluchanskiy A, Hedrick AF, Awasthi V, Subramanian M, Csiszar A, Balasubramanian P. Chronic β3 adrenergic agonist treatment improves brain microvascular endothelial function and cognition in aged mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602747. [PMID: 39026792 PMCID: PMC11257558 DOI: 10.1101/2024.07.09.602747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Microvascular endothelial dysfunction, characterized by impaired neurovascular coupling, reduced glucose uptake, blood-brain barrier disruption, and microvascular rarefaction, plays a critical role in the pathogenesis of age-related vascular cognitive impairment (VCI). Emerging evidence points to non-cell autonomous mechanisms mediated by adverse circulating milieu (an increased ratio of pro-geronic to anti-geronic circulating factors) in the pathogenesis of endothelial dysfunction leading to impaired cerebral blood flow and cognitive decline in the aging population. In particular, age-related adipose dysfunction contributes, at least in part, to an unfavorable systemic milieu characterized by chronic hyperglycemia, hyperinsulinemia, dyslipidemia, and altered adipokine profile, which together contribute to microvascular endothelial dysfunction. Hence, in the present study, we aimed to test whether thermogenic stimulation, an intervention known to improve adipose and systemic metabolism by increasing cellular energy expenditure, could mitigate brain endothelial dysfunction and improve cognition in the aging population. Eighteen-month-old old C57BL/6J mice were treated with saline or CL (β3-adrenergic agonist) for 6 weeks followed by functional analysis to assess endothelial function and cognition. CL treatment improved neurovascular coupling responses and rescued brain glucose uptake in aged animals. In addition, CL treatment also attenuated blood-brain barrier leakage and associated neuroinflammation in the cortex of aged animals. More importantly, these beneficial changes in microvascular function translated to improved cognitive performance in radial arm water maze and Y-maze tests. Our results suggest that β3-adrenergic agonist treatment improves multiple aspects of brain microvascular endothelial function and can be potentially repurposed for treating age-associated cognitive decline.
Collapse
|
24
|
Wang L, Liu Y, Li K, Zhang W, Yuan Y, Ma K, Zhou F, Cheng Z, Geng J, Su Y, Guo Z, Blake GM, Cheng X, Liu Y, Engelke K, Vlug AG. Age and BMI have different effects on subcutaneous, visceral, liver, bone marrow, and muscle adiposity, as measured by CT and MRI. Obesity (Silver Spring) 2024; 32:1339-1348. [PMID: 38783517 DOI: 10.1002/oby.24040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE We analyzed quantitative computed tomography (CT) and chemical shift-encoded magnetic resonance imaging (MRI) data from a Chinese cohort to investigate the effects of BMI and aging on different adipose tissue (AT) depots. METHODS In 400 healthy, community-dwelling individuals aged 22 to 83 years, we used MRI to quantify proton density fat fraction (PDFF) of the lumbar spine (L2-L4) bone marrow AT (BMAT), the psoas major and erector spinae (ES) muscles, and the liver. Abdominal total AT, visceral AT (VAT), and subcutaneous AT (SAT) areas were measured at the L2-L3 level using quantitative CT. Partial correlation analysis was used to evaluate the relationship of each AT variable with age and BMI. Multiple linear regression analysis was performed in which each AT variable was evaluated in turn as a function of age and the other five independent AT measurements. RESULTS Of the 168 men, 29% had normal BMI (<24.0 kg/m2), 47% had overweight (24.0-27.9 kg/m2), and 24% had obesity (≥ 28.0 kg/m2). In the 232 women, the percentages were 46%, 32%, and 22%, respectively. Strong or very strong correlations with BMI were found for total AT, VAT, and SAT in both sexes. BMAT and ES PDFF was strongly correlated with age in women and moderately correlated in men. In both sexes, BMAT PDFF correlated only with age and not with any of the other AT depots. Psoas PDFF correlated only with ES PDFF and not with age or the other AT depots. Liver PDFF correlated with BMI and VAT and weakly with SAT in men. VAT and SAT correlated with age and each other in both sexes. CONCLUSIONS Age and BMI are both associated with adiposity, but their effects differ depending on the type of AT.
Collapse
Affiliation(s)
- Ling Wang
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
- JST Sarcopenia Research Centre, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Yandong Liu
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Kai Li
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Wenshuang Zhang
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Yi Yuan
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Kangkang Ma
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Fengyun Zhou
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Zitong Cheng
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Jian Geng
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Yongbin Su
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Zhe Guo
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Glen M Blake
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Xiaoguang Cheng
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Yajun Liu
- JST Sarcopenia Research Centre, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
- Department of Spine Surgery, Beijing Jishuitan Hospital, Capital Medical University, National Center for Orthopaedics, Beijing, China
| | - Klaus Engelke
- Institute of Medical Physics, University Erlangen-Nuremberg, Nuremberg, Germany
- Department of Medicine 3 - Rheumatology and Immunology, FAU University of Erlangen-Nuremberg and University Hospital Erlangen, Erlangen, Germany
| | - Annegreet G Vlug
- Center for Bone Quality, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
25
|
Yuan Y, Hu R, Park J, Xiong S, Wang Z, Qian Y, Shi Z, Wu R, Han Z, Ong SG, Lin S, Varady KA, Xu P, Berry DC, Shu G, Jiang Y. Macrophage-derived chemokine CCL22 establishes local LN-mediated adaptive thermogenesis and energy expenditure. SCIENCE ADVANCES 2024; 10:eadn5229. [PMID: 38924414 PMCID: PMC11204298 DOI: 10.1126/sciadv.adn5229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
There is a regional preference around lymph nodes (LNs) for adipose beiging. Here, we show that local LN removal within inguinal white adipose tissue (iWAT) greatly impairs cold-induced beiging, and this impairment can be restored by injecting M2 macrophages or macrophage-derived C-C motif chemokine (CCL22) into iWAT. CCL22 injection into iWAT effectively promotes iWAT beiging, while blocking CCL22 with antibodies can prevent it. Mechanistically, the CCL22 receptor, C-C motif chemokine receptor 4 (CCR4), within eosinophils and its downstream focal adhesion kinase/p65/interleukin-4 signaling are essential for CCL22-mediated beige adipocyte formation. Moreover, CCL22 levels are inversely correlated with body weight and fat mass in mice and humans. Acute elevation of CCL22 levels effectively prevents diet-induced body weight and fat gain by enhancing adipose beiging. Together, our data identify the CCL22-CCR4 axis as an essential mediator for LN-controlled adaptive thermogenesis and highlight its potential to combat obesity and its associated complications.
Collapse
Affiliation(s)
- Yexian Yuan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ruoci Hu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jooman Park
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Shaolei Xiong
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zilai Wang
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yanyu Qian
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zuoxiao Shi
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ruifan Wu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenbo Han
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Shuhao Lin
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Krista A. Varady
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
26
|
Tang Q, Xing X, Huang H, Yang J, Li M, Xu X, Gao X, Liang C, Tian W, Liao L. Eliminating senescent cells by white adipose tissue-targeted senotherapy alleviates age-related hepatic steatosis through decreasing lipolysis. GeroScience 2024; 46:3149-3167. [PMID: 38217637 PMCID: PMC11009221 DOI: 10.1007/s11357-024-01068-5] [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: 07/11/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024] Open
Abstract
Cellular senescence is an important risk factor in the development of hepatic steatosis. Senolytics present therapeutic effects on age-related hepatic steatosis without eliminating senescent hepatocytes directly. Therefore, it highlights the need to find senolytics' therapeutic targets. Dysfunction of adipose tissue underlies the critical pathogenesis of lipotoxicity in the liver. However, the correlation between adipose tissue and hepatic steatosis during aging and its underlying molecular mechanism remains poorly understood. We explored the correlation between white adipose tissue (WAT) and the liver during aging and evaluated the effect of lipolysis of aged WAT on hepatic steatosis and hepatocyte senescence. We screened out the ideal senolytics for WAT and developed a WAT-targeted delivery system for senotherapy. We assessed senescence and lipolysis of WAT and hepatic lipid accumulation after treatment. The results displayed that aging accelerated cellular senescence and facilitated lipolysis of WAT. Free fatty acids (FFAs) generated by WAT during aging enhanced hepatic steatosis and induced hepatocyte senescence. The combined usage of dasatinib and quercetin was screened out as the ideal senolytics to eliminate senescent cells in WAT. To minimize non-specific distribution and enhance the effectiveness of senolytics, liposomes decorated with WAT affinity peptide P3 were constructed for senotherapy in vivo. In vivo study, WAT-targeted treatment eliminated senescent cells in WAT and reduced lipolysis, resulting in the alleviation of hepatic lipid accumulation and hepatocyte senescence when compared to non-targeted treatment, providing a novel tissue-targeted, effective and safe senotherapy for age-related hepatic steatosis.
Collapse
Affiliation(s)
- Qi Tang
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
| | - Xiaotao Xing
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Laboratory Center of Stomatology, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Haisen Huang
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
| | - Jian Yang
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
| | - Maojiao Li
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
| | - Xun Xu
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
| | - Xin Gao
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
| | - Cheng Liang
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China
| | - Weidong Tian
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| | - Li Liao
- National Engineering Laboratory for Oral Regenerative Medicine & Engineering Research Center of Oral Translational Medicine, Ministry of Education & State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, West China School of Public Health & West China Fourth Hospital, Sichuan University, No.14, 3Rd Section Of Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
27
|
de Oliveira RF, Salazar M, Matos L, Almeida H, Rodrigues AR, Gouveia AM. High copper levels induce premature senescence in 3T3-L1 preadipocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119734. [PMID: 38642724 DOI: 10.1016/j.bbamcr.2024.119734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/22/2024]
Abstract
Copper (Cu) dyshomeostasis has been linked to obesity and related morbidities and also to aging. Cu levels are higher in older or obese individuals, and adipose tissue (AT) Cu levels correlate with body mass index. Aging and obesity induce similar AT functional and structural changes, including an accumulation of senescent cells. To study the effect of Cu-mediated stress-induced premature senescent (Cu-SIPS) on preadipocytes, 3T3-L1 cell line was exposed to a subcytotoxic concentration of copper sulfate. After Cu treatment, preadipocytes acquired typical senescence characteristics including diminished cell proliferation, cell and nuclei enlargement and increased lysosomal mass (higher Lamp2 expression and a slight increased number of cells positive for β-galactosidase associated with senescence (SA-β-Gal)). Cell cycle arrest was due to upregulation of p16Ink4aInk4a and p21Waf1/Cip1. Accordingly, protein levels of the proliferation marker KI67 were reduced. Cu-SIPS relates with oxidative stress and, in this context, an increase of SOD1 and HO-1 expression was detected in Cu-treated cells. The mRNA expression of senescence-associated secretory phenotype factors, such as Mmp3, Il-6 and Tnf-α, increased in Cu-SIPS 3T3-L1 cells but no effect was observed on the expression of heterochromatin-associated protein 1(HP1). Although the downregulation of Lamin B1 expression is considered a hallmark of senescence, Cu-SIPS cells presented higher levels of Lamin B1. The dysregulation of nuclear lamina was accompanied by an increase of nuclear blebbing, but not of micronuclei number. To conclude, a Cu-SIPS model in 3T3-L1 preadipocytes is here described, which may be an asset to the study of AT dysregulation observed in obesity and aging.
Collapse
Affiliation(s)
- Ricardo F de Oliveira
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Maria Salazar
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Liliana Matos
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Faculdade de Ciências da Nutrição e Alimentação, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Henrique Almeida
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Adriana R Rodrigues
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Faculdade de Ciências da Nutrição e Alimentação, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Alexandra M Gouveia
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
28
|
Chu LX, Wang WJ, Gu XP, Wu P, Gao C, Zhang Q, Wu J, Jiang DW, Huang JQ, Ying XW, Shen JM, Jiang Y, Luo LH, Xu JP, Ying YB, Chen HM, Fang A, Feng ZY, An SH, Li XK, Wang ZG. Spatiotemporal multi-omics: exploring molecular landscapes in aging and regenerative medicine. Mil Med Res 2024; 11:31. [PMID: 38797843 PMCID: PMC11129507 DOI: 10.1186/s40779-024-00537-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Aging and regeneration represent complex biological phenomena that have long captivated the scientific community. To fully comprehend these processes, it is essential to investigate molecular dynamics through a lens that encompasses both spatial and temporal dimensions. Conventional omics methodologies, such as genomics and transcriptomics, have been instrumental in identifying critical molecular facets of aging and regeneration. However, these methods are somewhat limited, constrained by their spatial resolution and their lack of capacity to dynamically represent tissue alterations. The advent of emerging spatiotemporal multi-omics approaches, encompassing transcriptomics, proteomics, metabolomics, and epigenomics, furnishes comprehensive insights into these intricate molecular dynamics. These sophisticated techniques facilitate accurate delineation of molecular patterns across an array of cells, tissues, and organs, thereby offering an in-depth understanding of the fundamental mechanisms at play. This review meticulously examines the significance of spatiotemporal multi-omics in the realms of aging and regeneration research. It underscores how these methodologies augment our comprehension of molecular dynamics, cellular interactions, and signaling pathways. Initially, the review delineates the foundational principles underpinning these methods, followed by an evaluation of their recent applications within the field. The review ultimately concludes by addressing the prevailing challenges and projecting future advancements in the field. Indubitably, spatiotemporal multi-omics are instrumental in deciphering the complexities inherent in aging and regeneration, thus charting a course toward potential therapeutic innovations.
Collapse
Affiliation(s)
- Liu-Xi Chu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wen-Jia Wang
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xin-Pei Gu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
- Department of Human Anatomy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Ping Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Chen Gao
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Quan Zhang
- Integrative Muscle Biology Laboratory, Division of Regenerative and Rehabilitative Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Jia Wu
- Key Laboratory for Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Da-Wei Jiang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jun-Qing Huang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China
| | - Xin-Wang Ying
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jia-Men Shen
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi Jiang
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Li-Hua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 324025, Zhejiang, China
| | - Jun-Peng Xu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi-Bo Ying
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hao-Man Chen
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ao Fang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zun-Yong Feng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore.
| | - Shu-Hong An
- Department of Human Anatomy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Xiao-Kun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Zhou-Guang Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China.
| |
Collapse
|
29
|
Behtaj D, Ghorbani A, Eslamian G, Malekpour Alamdari N, Abbasi M, Zand H, Shakery A, Shimi G, Sohouli MH, Fazeli Taherian S. Ex vivo Anti-Senescence Activity of N-Acetylcysteine in Visceral Adipose Tissue of Obese Volunteers. Obes Facts 2024; 17:355-363. [PMID: 38718763 PMCID: PMC11299969 DOI: 10.1159/000539255] [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: 05/08/2023] [Accepted: 05/06/2024] [Indexed: 08/07/2024] Open
Abstract
INTRODUCTION Excessive visceral adiposity is known to drive the onset of metabolic derangements, mostly involving oxidative stress, prolonged inflammation, and cellular senescence. N-acetylcysteine (NAC) is a synthetic form of l-cysteine with potential antioxidant, anti-inflammatory, and anti-senescence properties. This ex-vivo study aimed to determine the effect of NAC on some markers of senescence including β-galactosidase activity and p16, p53, p21, IL-6, and TNF-α gene expressions in visceral adipose tissue in obese adults. METHODS This ex-vivo experimental study involved 10 obese participants who were candidates for bariatric surgery. Duplicate biopsies from the abdominal visceral adipose tissue were obtained from the omentum. The biopsies were treated with or without NAC (5 and 10 mm). To evaluate adipose tissue senescence, beta-galactosidase (β-gal) activity and the expression of P16, P21, P53, IL-6, and TNF-α were determined. ANOVA test was employed to analyze the varying markers of cellular senescence and inflammation between treatment groups. RESULTS The NAC at concentrations of 5 mm and 10 mm resulted in a noteworthy reduction β-gal activity compared to the control group (p < 0.001). Additionally, the expression of P16, P21, and IL-6 was significantly reduced following treatment with NAC (5 mm) and NAC (10 mm) compared to the control group (All p < 0.001). DISCUSSION/CONCLUSION Taken together, these data suggest the senotherapeutic effect of NAC, as it effectively reduces the activity of SA-β-gal and the expression of IL-6, P16, and P21 genes in the visceral adipose tissue of obese individuals.
Collapse
Affiliation(s)
- Diba Behtaj
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Ghorbani
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Eslamian
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Malekpour Alamdari
- School of Medicine, Department of General Surgery, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Abbasi
- School of Medicine, Department of General Surgery, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Zand
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Shakery
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Shimi
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hasan Sohouli
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Fazeli Taherian
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Chinnapaka S, Malekzadeh H, Tirmizi Z, Ejaz A. Caloric restriction mitigates age-associated senescence characteristics in subcutaneous adipose tissue-derived stem cells. Aging (Albany NY) 2024; 16:7535-7552. [PMID: 38728252 PMCID: PMC11131987 DOI: 10.18632/aging.205812] [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: 09/19/2023] [Accepted: 02/27/2024] [Indexed: 05/12/2024]
Abstract
Adipose tissue regulates metabolic balance, but aging disrupts it, shifting fat from insulin-sensitive subcutaneous to insulin-resistant visceral depots, impacting overall metabolic health. Adipose-derived stem cells (ASCs) are crucial for tissue regeneration, but aging diminishes their stemness and regeneration potential. Our findings reveal that aging is associated with a decrease in subcutaneous adipose tissue mass and an increase in the visceral fat depots mass. Aging is associated with increase in adipose tissue fibrosis but no significant change in adipocyte size was observed with age. Long term caloric restriction failed to prevent fibrotic changes but resulted in significant decrease in adipocytes size. Aged subcutaneous ASCs displayed an increased production of ROS. Using mitochondrial membrane activity as an indicator of stem cell quiescence and senescence, we observed a significant decrease in quiescence ASCs with age exclusively in subcutaneous adipose depot. In addition, aged subcutaneous adipose tissue accumulated more senescent ASCs having defective autophagy activity. However, long-term caloric restriction leads to a reduction in mitochondrial activity in ASCs. Furthermore, caloric restriction prevents the accumulation of senescent cells and helps retain autophagy activity in aging ASCs. These results suggest that caloric restriction and caloric restriction mimetics hold promise as a potential strategy to rejuvenate the stemness of aged ASCs. Further investigations, including in vivo evaluations using controlled interventions in animals and human studies, will be necessary to validate these findings and establish the clinical potential of this well-established approach for enhancing the stemness of aged stem cells.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hamid Malekzadeh
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zayaan Tirmizi
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
31
|
Zhang G, Samarawickrama PN, Gui L, Ma Y, Cao M, Zhu H, Li W, Yang H, Li K, Yang Y, Zhu E, Li W, He Y. Revolutionizing Diabetic Foot Ulcer Care: The Senotherapeutic Approach. Aging Dis 2024; 16:946-970. [PMID: 38739931 PMCID: PMC11964433 DOI: 10.14336/ad.2024.0065] [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: 01/17/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are a prevalent and profoundly debilitating complication that afflicts individuals with diabetes mellitus (DM). These ulcers are associated with substantial morbidity, recurrence rates, disability, and mortality, imposing substantial economic, psychological, and medical burdens. Timely detection and intervention can mitigate the morbidity and disparities linked to DFU. Nevertheless, current therapeutic approaches for DFU continue to grapple with multifaceted limitations. A growing body of evidence emphasizes the crucial role of cellular senescence in the pathogenesis of chronic wounds. Interventions that try to delay cellular senescence, eliminate senescent cells (SnCs), or suppress the senescence-associated secretory phenotype (SASP) have shown promise for helping chronic wounds to heal. In this context, targeting cellular senescence emerges as a novel therapeutic strategy for DFU. In this comprehensive review, we look at the pathology and treatment of DFU in a systematic way. We also explain the growing importance of investigating SnCs in DFU and highlight the great potential of senotherapeutics that target SnCs in DFU treatment. The development of efficacious and safe senotherapeutics represents a pioneering therapeutic approach aimed at enhancing the quality of life for individuals affected by DFU.
Collapse
Affiliation(s)
- Guiqin Zhang
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Priyadarshani Nadeeshika Samarawickrama
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| | - Li Gui
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Yuan Ma
- Department of Orthopedics, the Third People’s Hospital of Yunnan Province, Kunming, Yunnan 650011, China.
| | - Mei Cao
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Hong Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Wei Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Honglin Yang
- Department of Orthopedics, the Third People’s Hospital of Yunnan Province, Kunming, Yunnan 650011, China.
| | - Kecheng Li
- Department of Orthopedics, the Third People’s Hospital of Yunnan Province, Kunming, Yunnan 650011, China.
| | - Yang Yang
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | - Enfang Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Wen Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Yonghan He
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| |
Collapse
|
32
|
Yu L, Wan Q, Liu Q, Fan Y, Zhou Q, Skowronski AA, Wang S, Shao Z, Liao CY, Ding L, Kennedy BK, Zha S, Que J, LeDuc CA, Sun L, Wang L, Qiang L. IgG is an aging factor that drives adipose tissue fibrosis and metabolic decline. Cell Metab 2024; 36:793-807.e5. [PMID: 38378001 PMCID: PMC11070064 DOI: 10.1016/j.cmet.2024.01.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/31/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024]
Abstract
Aging is underpinned by pronounced metabolic decline; however, the drivers remain obscure. Here, we report that IgG accumulates during aging, particularly in white adipose tissue (WAT), to impair adipose tissue function and metabolic health. Caloric restriction (CR) decreases IgG accumulation in WAT, whereas replenishing IgG counteracts CR's metabolic benefits. IgG activates macrophages via Ras signaling and consequently induces fibrosis in WAT through the TGF-β/SMAD pathway. Consistently, B cell null mice are protected from aging-associated WAT fibrosis, inflammation, and insulin resistance, unless exposed to IgG. Conditional ablation of the IgG recycling receptor, neonatal Fc receptor (FcRn), in macrophages prevents IgG accumulation in aging, resulting in prolonged healthspan and lifespan. Further, targeting FcRn by antisense oligonucleotide restores WAT integrity and metabolic health in aged mice. These findings pinpoint IgG as a hidden culprit in aging and enlighten a novel strategy to rejuvenate metabolic health.
Collapse
Affiliation(s)
- Lexiang Yu
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Qianfen Wan
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Qiongming Liu
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Yong Fan
- Naomi Berrie Diabetes Center, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Alicja A Skowronski
- Naomi Berrie Diabetes Center, Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Summer Wang
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Zhengping Shao
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Chen-Yu Liao
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Lei Ding
- Department of Rehabilitation and Regenerative Medicine, Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brian K Kennedy
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Healthy Longevity Translational Research Programme, Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Health Longevity, National University Health System, Singapore, Singapore
| | - Shan Zha
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Jianwen Que
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Charles A LeDuc
- Naomi Berrie Diabetes Center, Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Liheng Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Department of Medicine, Division of Endocrinology, Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Li Qiang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Naomi Berrie Diabetes Center, Department of Medicine, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
33
|
Xiong L, Zhevlakova I, West XZ, Gao D, Murtazina R, Horak A, Brown JM, Molokotina I, Podrez EA, Byzova TV. TLR2 regulates hair follicle cycle and regeneration via BMP signaling. eLife 2024; 12:RP89335. [PMID: 38483447 PMCID: PMC10939499 DOI: 10.7554/elife.89335] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024] Open
Abstract
The etiology of hair loss remains enigmatic, and current remedies remain inadequate. Transcriptome analysis of aging hair follicles uncovered changes in immune pathways, including Toll-like receptors (TLRs). Our findings demonstrate that the maintenance of hair follicle homeostasis and the regeneration capacity after damage depend on TLR2 in hair follicle stem cells (HFSCs). In healthy hair follicles, TLR2 is expressed in a cycle-dependent manner and governs HFSCs activation by countering inhibitory BMP signaling. Hair follicles in aging and obesity exhibit a decrease in both TLR2 and its endogenous ligand carboxyethylpyrrole (CEP), a metabolite of polyunsaturated fatty acids. Administration of CEP stimulates hair regeneration through a TLR2-dependent mechanism. These results establish a novel connection between TLR2-mediated innate immunity and HFSC activation, which is pivotal to hair follicle health and the prevention of hair loss and provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Luyang Xiong
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Irina Zhevlakova
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Xiaoxia Z West
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Rakhilya Murtazina
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Anthony Horak
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - J Mark Brown
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Iuliia Molokotina
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| |
Collapse
|
34
|
Linders AN, Dias IB, López Fernández T, Tocchetti CG, Bomer N, Van der Meer P. A review of the pathophysiological mechanisms of doxorubicin-induced cardiotoxicity and aging. NPJ AGING 2024; 10:9. [PMID: 38263284 PMCID: PMC10806194 DOI: 10.1038/s41514-024-00135-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
The population of cancer survivors is rapidly increasing due to improving healthcare. However, cancer therapies often have long-term side effects. One example is cancer therapy-related cardiac dysfunction (CTRCD) caused by doxorubicin: up to 9% of the cancer patients treated with this drug develop heart failure at a later stage. In recent years, doxorubicin-induced cardiotoxicity has been associated with an accelerated aging phenotype and cellular senescence in the heart. In this review we explain the evidence of an accelerated aging phenotype in the doxorubicin-treated heart by comparing it to healthy aged hearts, and shed light on treatment strategies that are proposed in pre-clinical settings. We will discuss the accelerated aging phenotype and the impact it could have in the clinic and future research.
Collapse
Affiliation(s)
- Annet Nicole Linders
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Itamar Braga Dias
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Teresa López Fernández
- Division of Cardiology, Cardiac Imaging and Cardio-Oncology Unit, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Federico II University, Naples, Italy
- Centre for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
- Interdepartmental Centre of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Centre (CIRIAPA), Federico II University, Naples, Italy
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Peter Van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands.
| |
Collapse
|
35
|
Choi Y, Kim D, Kim SK. Effects of Physical Activity on Body Composition, Muscle Strength, and Physical Function in Old Age: Bibliometric and Meta-Analyses. Healthcare (Basel) 2024; 12:197. [PMID: 38255085 PMCID: PMC10815094 DOI: 10.3390/healthcare12020197] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVES Accumulating evidence suggests that physical activity (PA) is an efficient intervention to maintain functional capabilities and mitigate physiological changes in the older population. However, an attempt has yet to be made to comprehensively investigate the published landscape on the subject. METHODS This study had two aims. The first aim was to perform a bibliometric analysis for two keywords, "aging" and "PA", to analyze the research trend. Since "frailty" was the most noticeable co-occurring keyword with the two keywords, the second aim was to investigate the effects of PA, particularly, resistance training (RT), on frailty using a meta-analysis to provide a summary of the current evidence base. RESULTS The bibliometric analysis revealed that the number of publications on this research topic has gradually increased, highlighting the importance of understanding the role of PA in aging. The meta-analysis found that RT had significant beneficial effects on physical frailty factors, including handgrip strength, lower limb strength, balance, gait speed, and stair-climbing ability. CONCLUSION These findings demonstrate that RT is an effective intervention for improving physical function in frail populations; thus, it has important implications for the development of PA programs for older adults with frailty. Future research is warranted to explore the optimal dose, frequency, and duration of RT programs for older adults, as well as the potential benefits of combining RT with other forms of PA, such as aerobic or balance exercises.
Collapse
Affiliation(s)
- Yerim Choi
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea;
| | - Daekyoo Kim
- Department of Physical Education, Korea University, Seoul 02841, Republic of Korea;
| | - Seung Kyum Kim
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea;
- Department of Sports Science, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
36
|
Yang X, Xue C, Chen K, Gao D, Wang H, Tang C. Characteristics of elderly diabetes patients: focus on clinical manifestation, pathogenic mechanism, and the role of traditional Chinese medicine. Front Pharmacol 2024; 14:1339744. [PMID: 38273819 PMCID: PMC10808572 DOI: 10.3389/fphar.2023.1339744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Diabetes mellitus has become a major public health issue globally, putting an enormous burden on global health systems and people. Among all diseased groups, a considerable part of patients are elderly, while their clinical features, pathogenic processes, and medication regimens are different from patients of other ages. Despite the availability of multiple therapies and techniques, there are still numerous elderly diabetes patients suffering from poor blood glucose control, severe complications, and drug adverse effects, which negatively affect the quality of life in their golden years. Traditional Chinese Medicine (TCM) has been widely used in the treatment of diabetes for several decades, and its relevant clinical practice has confirmed that it has a satisfactory effect on alleviating clinical symptoms and mitigating the progression of complications. Chinese herbal medicine and its active components were used widely with obvious clinical advantages by multiple targets and signaling pathways. However, due to the particular features of elderly diabetes, few studies were conducted to explore Traditional Chinese Medicine intervention on elderly diabetic patients. This study reviews the research on clinical features, pathogenic processes, treatment principles, and TCM treatments, hoping to provide fresh perspectives on the prevention and management strategies for elderly diabetes.
Collapse
Affiliation(s)
- Xiaofei Yang
- Beijing University of Chinese Medicine, Beijing, China
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chongxiang Xue
- Beijing University of Chinese Medicine, Beijing, China
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Keyu Chen
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dongyang Gao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Han Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Cheng Tang
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
37
|
Mukherjee S, Bruno MEC, Oakes J, Hawk GS, Stromberg AJ, Cohen DA, Starr ME. Mechanisms of γδ T cell accumulation in visceral adipose tissue with aging. FRONTIERS IN AGING 2024; 4:1258836. [PMID: 38274288 PMCID: PMC10808514 DOI: 10.3389/fragi.2023.1258836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024]
Abstract
γδ T cells are resident in visceral adipose tissue (VAT) where they show an age-associated increase in numbers and contribute to local and systemic chronic inflammation. However, regulation of this population and mechanisms for the age-dependent accumulation are not known. In this study, we identified a progressive trend of γδ T cell accumulation in VAT over the lifespan in mice and explored physiological mechanisms contributing to accumulation. Using isochronic parabiotic pairs of wild-type (WT) and T cell receptor delta knockout (TCRδ KO) mice at young and old age, we confirmed that VAT γδ T cells are predominately a tissue-resident population which is sustained in aging. Migration of peripheral γδ T cells into VAT was observed at less than 10%, with a decreasing trend by aging, suggesting a minor contribution of recruitment to γδ T cell accumulation with aging. Since tissue-resident T cell numbers are tightly regulated by a balance between proliferation and programmed cell death, we further explored these processes. Using in vivo EdU incorporation and the proliferation marker Ki67, we found that the absolute number of proliferating γδ T cells in VAT is significantly higher in the aged compared to young and middle-aged mice, despite a decline in the proportion of proliferating to non-proliferating cells by age. Analysis of apoptosis via caspase 3/7 activation revealed that VAT γδ T cells show reduced apoptosis starting at middle age and continuing into old age. Further, induction of apoptosis using pharmacological inhibitors of Bcl2 family proteins revealed that VAT γδ T cells at middle age are uniquely protected from apoptosis via a mechanism independent of traditional anti-apoptotic Bcl2-family proteins. Collectively, these data indicate that protection from apoptosis at middle age increases survival of tissue-resident γδ T cells resulting in an increased number of proliferative cells from middle age onward, and leading to the age-associated accumulation of γδ T cells in VAT. These findings are important to better understand how adipose tissue dysfunction and related changes in the immune profile contribute to inflammaging among the elderly.
Collapse
Affiliation(s)
- Sujata Mukherjee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Maria E. C. Bruno
- Division of Research, Department of Surgery, University of Kentucky, Lexington, KY, United States
| | - Jason Oakes
- Division of Laboratory Animal Resources, University of Kentucky, Lexington, KY, United States
| | - Gregory S. Hawk
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Arnold J. Stromberg
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Donald A. Cohen
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Marlene E. Starr
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
- Division of Research, Department of Surgery, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
38
|
Kordi N, Saydi A, Karami S, Bagherzadeh-Rahmani B, Marzetti E, Jung F, Stockwell BR. Ferroptosis and aerobic training in ageing. Clin Hemorheol Microcirc 2024; 87:347-366. [PMID: 38306027 DOI: 10.3233/ch-232076] [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: 02/03/2024]
Abstract
Ferroptosis is a form of programmed cell death that plays a significant role in causing several diseases such as heart attack and heart failure, through alterations in fat, amino acid, and iron metabolism. Comprehending the regulatory mechanisms of ferroptosis signaling is critical because it has a considerable effect on the elderly's mortality. Conversely, age-related changes in substrate metabolism and metabolite levels are recognized to give rise to obesity. Furthermore, research has proposed that aging and obesity-related changes in substrate metabolism may aggravate ferroptosis. The suppression of ferroptosis holds potential as a successful therapeutic approach for managing different diseases, including sarcopenia, cardiovascular diseases, and central nervous system diseases. However, the pathologic and biological mechanisms behind the function of ferroptosis are not fully comprehended yet. Physical activity could affect lipid, amino acid, and iron metabolism to modulate ferroptosis. The aim of this study is to showcase the current understanding of the molecular mechanisms leading to ferroptosis and discuss the role of aging and physical activity in this phenomenon.
Collapse
Affiliation(s)
- Negin Kordi
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Ali Saydi
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Sajad Karami
- Faculty of Physical Education and Sport Science, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Behnam Bagherzadeh-Rahmani
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Emanuele Marzetti
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Friedrich Jung
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, NewYork, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
| |
Collapse
|
39
|
Lan R, Wei L, Yu H, Jiang P, Zhao Z. Age-Related Changes in Hepatic Lipid Metabolism and Abdominal Adipose Deposition in Yellow-Feathered Broilers Aged from 1 to 56 Days. Animals (Basel) 2023; 13:3860. [PMID: 38136897 PMCID: PMC10740587 DOI: 10.3390/ani13243860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The objective of this study was to evaluate the age-related changes in hepatic lipid metabolism, adipocyte hyperplasia, hypertrophy, and lipid metabolism in the abdominal adipose tissue of yellow-feathered broilers. Blood, liver, and abdominal adipose samples were collected on days 1, 7, 14, 21, 28, 35, 42, 49, and 56. Body, liver, and abdominal weight increased (p < 0.05) with age-related changes. The triacylglycerol content peaked on day 14, and total cholesterol content peaked on day 56. The adipocyte diameter and area peaked on day 56, and total DNA content peaked on day 7. The age-related changes in hepatic lipogenesis-related gene (ChREBP, SREBP-1c, ACC, FAS, SCD1) expression mainly occurred during days 1 to 21, hepatic lipolysis-related gene (CPT1, LPL, ApoB) expression mainly occurred during days 1 to 14, and abdominal adipose-deposition-related gene (PPARα, CPT1, LPL, PPARγ, C/EBPβ) expression occurred during days 1 to 14. These results demonstrated a dynamic pattern of hepatic lipid metabolism and abdominal adipose deposition in yellow-feathered broilers, which provides practical strategies to regulate hepatic lipid metabolism and reduce abdominal adipose deposition in yellow-feathered broilers.
Collapse
Affiliation(s)
| | | | | | | | - Zhihui Zhao
- Department of Animal Science and Technology, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (R.L.); (L.W.); (H.Y.); (P.J.)
| |
Collapse
|
40
|
Bilibio BLE, Dos Reis WR, Compagnon L, de Batista DG, Sulzbacher LM, Pinheiro JF, Ludwig MS, Frizzo MN, Cruzat V, Heck TG. Effects of alternate-day fasting and time-restricted feeding in obese middle-aged female rats. Nutrition 2023; 116:112198. [PMID: 37717500 DOI: 10.1016/j.nut.2023.112198] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/12/2023] [Accepted: 08/13/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVES Obesity is a multifactorial condition associated with metabolic alterations that can be aggravated during female aging. Calorie restriction via intermittent fasting (IF) diets may reduce body weight and therefore have the potential to decrease obesity and associated comorbidities, such as insulin resistance. This study investigated the effects of two IF protocols, alternate-day fasting (ADF) and time-restricted feeding (TRF) in middle-aged obese female rats. METHODS Wistar rats (age 15 mo) were fed with standard chow or high-fat diet for 8 wk and then separated into the following groups (n = 5-8 each) for another 8 wk: control (received standard chow), obese (received high-fat diet), obese + ADF (24-h fasting protocol), and obese + TRF (14 h daily). RESULTS At the end of the study, both IF protocols were able to reduce body weight and body mass index compared with the obese group. However, no changes were observed in adiposity and glucose homeostasis. We also found an increase in total leukocytes, lymphocytes, and monocytes in the TRF group and a higher number of platelets in the ADF group. Blood lipid profiles, including triglycerides and high-density lipoprotein, as well as liver stress responses, such as heat shock protein 70 and malondialdehyde, were not changed by IF. CONCLUSIONS Although ADF and TRF protocols resulted in a reduction of body weight and body mass index, these dietary interventions did not promote health benefits, such as reducing blood lipid profile, adiposity, and insulin resistance. In addition, ADF and TRF increased inflammatory biomarkers, which may increase the risk of obesity-associated comorbidities.
Collapse
Affiliation(s)
- Bruna L Endl Bilibio
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Welerson R Dos Reis
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Letícia Compagnon
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Diovana G de Batista
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Mathematical and Computational Modelling, Regional University of North-western Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Lucas M Sulzbacher
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Juliana F Pinheiro
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Mirna S Ludwig
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Matias N Frizzo
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| | - Vinicius Cruzat
- Faculty of Health, Southern Cross University, Gold Coast, Queensland, Australia.
| | - Thiago G Heck
- Research Group in Physiology, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil; Postgraduate Program in Mathematical and Computational Modelling, Regional University of North-western Rio Grande do Sul State, Ijuí, Rio Grande do Sul State, Brazil
| |
Collapse
|
41
|
Bauer C, Sim M, Prince RL, Zhu K, Lim EM, Byrnes E, Pavlos N, Lim WH, Wong G, Lewis JR, Levinger I. Circulating lipocalin-2 and features of metabolic syndrome in community-dwelling older women: A cross-sectional study. Bone 2023; 176:116861. [PMID: 37524293 DOI: 10.1016/j.bone.2023.116861] [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: 04/10/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Lipocalin-2 (LCN2) is released by several cell types including osteoblasts and adipocytes and has been suggested as a marker of renal dysfunction, metabolic syndrome (MetS) and type 2 diabetes (T2D). Whether LCN2 is linked to these diseases in older women remains unknown. This study investigated whether LCN2 is related to features of MetS and T2D in older women. This cross-sectional study included 705 non-diabetic women (mean age 75.1 ± 2.6 years) for MetS analysis and 76 women (mean age 75.4 ± 2.8 years) with T2D. Total circulating LCN2 levels were analysed using a two-step chemiluminescent microparticle monoclonal immunoassay. MetS was determined by a modified National Cholesterol Education Program Adult Treatment Panel III classification. Multivariable-adjusted logistic regression analysis was used to assess odds ratios between LCN2 quartiles and MetS. Women in the highest LCN2 quartile had approximately 3 times greater risk for MetS compared to women in the lowest quartile (OR 3.05; 95%CI 1.86-5.02). Women with T2D or MetS scores of ≥ 3 had higher LCN2 levels compared to women with a MetS score of 0 (p < 0.05). Higher LCN2 correlated with higher body mass index, fat mass, triglycerides and glycated haemoglobin and lower high-density lipoprotein cholesterol and estimated glomerular filtration rate (p < 0.05). Higher circulating levels of LCN2 are associated with worsened cardio-metabolic risk factors and increased odds of MetS and T2D in older women. Whether it can be used as a biomarker for identifying those at risk for MetS and T2D should be explored further.
Collapse
Affiliation(s)
- Carlie Bauer
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Marc Sim
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, Australia; Medical School, University of Western Australia, Perth, Australia
| | - Richard L Prince
- Medical School, University of Western Australia, Perth, Australia
| | - Kun Zhu
- Medical School, University of Western Australia, Perth, Australia; Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
| | - Ee M Lim
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia; PathWest, QEII Medical Centre, Perth, Australia
| | | | - Nathan Pavlos
- School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Wai H Lim
- Medical School, University of Western Australia, Perth, Australia; Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Germaine Wong
- Centre for Kidney Research, Children's Hospital at Westmead School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Joshua R Lewis
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, Australia; Medical School, University of Western Australia, Perth, Australia; Centre for Kidney Research, Children's Hospital at Westmead School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Itamar Levinger
- Institute for Health and Sport, Victoria University, Melbourne, Australia; Australian Institute for Musculoskeletal Science, Victoria University, University of Melbourne, Western Health, St Albans, VIC, Australia.
| |
Collapse
|
42
|
Grun LK, Maurmann RM, Scholl JN, Fogaça ME, Schmitz CRR, Dias CK, Gasparotto J, Padoin AV, Mottin CC, Klamt F, Figueiró F, Jones MH, Filippi-Chiela EC, Guma FCR, Barbé-Tuana FM. Obesity drives adipose-derived stem cells into a senescent and dysfunctional phenotype associated with P38MAPK/NF-KB axis. Immun Ageing 2023; 20:51. [PMID: 37821967 PMCID: PMC10566105 DOI: 10.1186/s12979-023-00378-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Adipose-derived stem cells (ADSC) are multipotent cells implicated in tissue homeostasis. Obesity represents a chronic inflammatory disease associated with metabolic dysfunction and age-related mechanisms, with progressive accumulation of senescent cells and compromised ADSC function. In this study, we aimed to explore mechanisms associated with the inflammatory environment present in obesity in modulating ADSC to a senescent phenotype. We evaluated phenotypic and functional alterations through 18 days of treatment. ADSC were cultivated with a conditioned medium supplemented with a pool of plasma from eutrophic individuals (PE, n = 15) or with obesity (PO, n = 14), and compared to the control. RESULTS Our results showed that PO-treated ADSC exhibited decreased proliferative capacity with G2/M cycle arrest and CDKN1A (p21WAF1/Cip1) up-regulation. We also observed increased senescence-associated β-galactosidase (SA-β-gal) activity, which was positively correlated with TRF1 protein expression. After 18 days, ADSC treated with PO showed augmented CDKN2A (p16INK4A) expression, which was accompanied by a cumulative nuclear enlargement. After 10 days, ADSC treated with PO showed an increase in NF-κB phosphorylation, while PE and PO showed an increase in p38MAPK activation. PE and PO treatment also induced an increase in senescence-associated secretory phenotype (SASP) cytokines IL-6 and IL-8. PO-treated cells exhibited decreased metabolic activity, reduced oxygen consumption related to basal respiration, increased mitochondrial depolarization and biomass, and mitochondrial network remodeling, with no superoxide overproduction. Finally, we observed an accumulation of lipid droplets in PO-treated ADSC, implying an adaptive cellular mechanism induced by the obesogenic stimuli. CONCLUSIONS Taken together, our data suggest that the inflammatory environment observed in obesity induces a senescent phenotype associated with p38MAPK/NF-κB axis, which stimulates and amplifies the SASP and is associated with impaired mitochondrial homeostasis.
Collapse
Affiliation(s)
- L K Grun
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| | - R M Maurmann
- Graduate Program in Cellular and Molecular Biology, School of Health, Sciences, and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - J N Scholl
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - M E Fogaça
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - C R R Schmitz
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - C K Dias
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - J Gasparotto
- Institute of Biomedical Sciences, Federal University at Alfenas, Alfenas, Brazil
| | - A V Padoin
- Graduate Program in Medicine and Health Sciences, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - C C Mottin
- Graduate Program in Medicine and Health Sciences, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - F Klamt
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - F Figueiró
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - M H Jones
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - E C Filippi-Chiela
- Institute of Basic Health Sciences, Department of Morphological Sciences, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Center for Biotechnology, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - F C R Guma
- Graduate Program in Biological Sciences: Biochemistry, Federal University at Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - F M Barbé-Tuana
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Graduate Program in Cellular and Molecular Biology, School of Health, Sciences, and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Group of Inflammation and Cellular Senescence, Immunobiology Laboratory, School of Health Sciences and Life, Pontifical Catholic University at Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
43
|
Xiong L, Zhevlakova I, West XZ, Gao D, Murtazina R, Horak A, Brown JM, Molokotina I, Podrez EA, Byzova TV. TLR2 Regulates Hair Follicle Cycle and Regeneration via BMP Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553236. [PMID: 37645905 PMCID: PMC10462054 DOI: 10.1101/2023.08.14.553236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The etiology of hair loss remains enigmatic, and current remedies remain inadequate. Transcriptome analysis of aging hair follicles uncovered changes in immune pathways, including Toll-like receptors (TLRs). Our findings demonstrate that the maintenance of hair follicle homeostasis and the regeneration capacity after damage depends on TLR2 in hair follicle stem cells (HFSCs). In healthy hair follicles, TLR2 is expressed in a cycle-dependent manner and governs HFSCs activation by countering inhibitory BMP signaling. Hair follicles in aging and obesity exhibit a decrease in both TLR2 and its endogenous ligand carboxyethylpyrrole (CEP), a metabolite of polyunsaturated fatty acids. Administration of CEP stimulates hair regeneration through a TLR2-dependent mechanism. These results establish a novel connection between TLR2-mediated innate immunity and HFSC activation, which is pivotal to hair follicle health and the prevention of hair loss and provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Luyang Xiong
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - Irina Zhevlakova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - Xiaoxia Z. West
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - Rakhylia Murtazina
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
- Current address: Department of Biochemistry and Molecular Genetics, University of Illinois; Chicago, IL 60607, USA
| | - Anthony Horak
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - J. Mark Brown
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - Iuliia Molokotina
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - Eugene A. Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| | - Tatiana V. Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic; Cleveland, OH 44195, USA
| |
Collapse
|
44
|
Chen D, Werder EJ, Stewart PA, Stenzel MR, Gerr FE, Lawrence KG, Groth CP, Huynh TB, Ramachandran G, Banerjee S, Jackson Ii WB, Christenbury K, Kwok RK, Sandler DP, Engel LS. Exposure to volatile hydrocarbons and neurologic function among oil spill workers up to 6 years after the Deepwater Horizon disaster. ENVIRONMENTAL RESEARCH 2023; 231:116069. [PMID: 37149022 PMCID: PMC10330421 DOI: 10.1016/j.envres.2023.116069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND During the 2010 Deepwater Horizon (DWH) disaster, oil spill response and cleanup (OSRC) workers were exposed to toxic volatile components of crude oil. Few studies have examined exposure to individual volatile hydrocarbon chemicals below occupational exposure limits in relation to neurologic function among OSRC workers. OBJECTIVES To investigate the association of several spill-related chemicals (benzene, toluene, ethylbenzene, xylene, n-hexane, i.e., BTEX-H) and total petroleum hydrocarbons (THC) with neurologic function among DWH spill workers enrolled in the Gulf Long-term Follow-up Study. METHODS Cumulative exposure to THC and BTEX-H across the oil spill cleanup period were estimated using a job-exposure matrix that linked air measurement data to detailed self-reported DWH OSRC work histories. We ascertained quantitative neurologic function data via a comprehensive test battery at a clinical examination that occurred 4-6 years after the DWH disaster. We used multivariable linear regression and modified Poisson regression to evaluate relationships of exposures (quartiles (Q)) with 4 neurologic function measures. We examined modification of the associations by age at enrollment (<50 vs. ≥50 years). RESULTS We did not find evidence of adverse neurologic effects from crude oil exposures among the overall study population. However, among workers ≥50 years of age, several individual chemical exposures were associated with poorer vibrotactile acuity of the great toe, with statistically significant effects observed in Q3 or Q4 of exposures (range of log mean difference in Q4 across exposures: 0.13-0.26 μm). We also observed suggestive adverse associations among those ≥ age 50 years for tests of postural stability and single-leg stance, although most effect estimates did not reach thresholds of statistical significance (p < 0.05). CONCLUSIONS Higher exposures to volatile components of crude oil were associated with modest deficits in neurologic function among OSRC workers who were age 50 years or older at study enrollment.
Collapse
Affiliation(s)
- Dazhe Chen
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Emily J Werder
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Mark R Stenzel
- Exposure Assessment Applications, LLC, Arlington, VA, USA
| | - Fredric E Gerr
- Department of Occupational and Environmental Health, University of Iowa College of Public Health, Iowa City, IA, USA
| | - Kaitlyn G Lawrence
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Caroline P Groth
- Department of Epidemiology and Biostatistics, School of Public Health, West Virginia University, Morgantown, WV, USA
| | - Tran B Huynh
- Department of Environmental and Occupational Health, Dornsife School of Public Health, Drexel University, Philadelphia, PA, USA
| | - Gurumurthy Ramachandran
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sudipto Banerjee
- Department of Biostatistics, Fielding School of Public Health, University of California - Los Angeles, Los Angeles, CA, USA
| | | | - Kate Christenbury
- Social & Scientific Systems, Inc, a DLH Holdings Company, Durham, NC, USA
| | - Richard K Kwok
- Population Studies and Genetics Branch, National Institute on Aging, Bethesda, MD, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Lawrence S Engel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA; Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
45
|
Chinnapaka S, Malekzadeh H, Tirmizi Z, Arellano JA, Ejaz A. Nicotinamide Riboside Improves Stemness of Human Adipose-Derived Stem Cells and Inhibits Terminal Adipocyte Differentiation. Pharmaceuticals (Basel) 2023; 16:1134. [PMID: 37631051 PMCID: PMC10458272 DOI: 10.3390/ph16081134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Adipose tissue plays a crucial role in maintaining metabolic homeostasis by serving as a storage site for excess fat and protecting other organs from the detrimental effects of lipotoxicity. However, the aging process is accompanied by a redistribution of fat, characterized by a decrease in insulin-sensitive subcutaneous adipose depot and an increase in insulin-resistant visceral adipose depot. This age-related alteration in adipose tissue distribution has implications for metabolic health. Adipose-derived stem cells (ASCs) play a vital role in the regeneration of adipose tissue. However, aging negatively impacts the stemness and regenerative potential of ASCs. The accumulation of oxidative stress and mitochondrial dysfunction-associated cellular damage contributes to the decline in stemness observed in aged ASCs. Nicotinamide adenine dinucleotide (NAD+) is a crucial metabolite that is involved in maintaining cellular homeostasis and stemness. The dysregulation of NAD+ levels with age has been associated with metabolic disorders and the loss of stemness. In this study, we aimed to investigate the effects of nicotinamide riboside (NR), a precursor of NAD+, on the stemness of human ASCs in cell culture. Our findings reveal that adipogenesis is accompanied by an increase in mitochondrial activity and the production of reactive oxygen species (ROS). However, treatment with NR leads to a reduction in mitochondrial activity and ROS production in ASCs. Furthermore, NR administration improves the stemness-related genes expression in ASCs and mitigates their propensity for adipocyte differentiation. These results suggest that NR treatment holds promise as a potential strategy to rejuvenate the stemness of aged ASCs. Further investigations, including in vivo evaluations using animal models and human studies, will be necessary to validate these findings and establish the clinical potential of this well-established drug for enhancing the stemness of aged stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
46
|
Zoico E, Saatchi T, Nori N, Mazzali G, Rizzatti V, Pizzi E, Fantin F, Giani A, Urbani S, Zamboni M. Senescent adipocytes as potential effectors of muscle cells dysfunction: An in vitro model. Exp Gerontol 2023; 179:112233. [PMID: 37321332 DOI: 10.1016/j.exger.2023.112233] [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: 11/03/2022] [Revised: 05/31/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Recently, there has been a growing body of evidence showing a negative effect of the white adipose tissue (WAT) dysfunction on the skeletal muscle function and quality. However, little is known about the effects of senescent adipocytes on muscle cells. Therefore, to explore potential mechanisms involved in age-related loss of muscle mass and function, we performed an in vitro experiment using conditioned medium obtained from cultures of mature and aged 3 T3-L1 adipocytes, as well as from cultures of dysfunctional adipocytes exposed to oxidative stress or high insulin doses, to treat C2C12 myocytes. The results from morphological measures indicated a significant decrease in diameter and fusion index of myotubes after treatment with medium of aged or stressed adipocytes. Aged and stressed adipocytes presented different morphological characteristics as well as a different gene expression profile of proinflammatory cytokines and ROS production. In myocytes treated with different adipocytes' conditioned media, we demonstrated a significant reduction of gene expression of myogenic differentiation markers as well as a significant increase of genes involved in atrophy. Finally, a significant reduction in protein synthesis as well as a significant increase of myostatin was found in muscle cells treated with medium of aged or stressed adipocytes compared to controls. In conclusion, these preliminary results suggest that aged adipocytes could influence negatively trophism, function and regenerative capacity of myocytes by a paracrine network of signaling.
Collapse
Affiliation(s)
- Elena Zoico
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Tanaz Saatchi
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy.
| | - Nicole Nori
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Gloria Mazzali
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Vanni Rizzatti
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| | - Eleonora Pizzi
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| | - Francesco Fantin
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Anna Giani
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Silvia Urbani
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Mauro Zamboni
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| |
Collapse
|
47
|
Serra M, Casas A, Teixeira JA, Barros AN. Revealing the Beauty Potential of Grape Stems: Harnessing Phenolic Compounds for Cosmetics. Int J Mol Sci 2023; 24:11751. [PMID: 37511513 PMCID: PMC10380576 DOI: 10.3390/ijms241411751] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Grape stems have emerged as a promising natural ingredient in the cosmetics industry due to their abundance of phenolic compounds, known for their antioxidant and anti-inflammatory properties. These compounds have shown great potential in promoting skin health, fighting signs of aging, and shielding against environmental stressors. With high concentrations of resveratrol, flavonoids, and tannins, grape stems have garnered attention from cosmetic scientists. Research has indicated that phenolic compounds extracted from grape stems possess potent antioxidant abilities, effectively combating free radicals that accelerate aging. Moreover, these compounds have demonstrated the capacity to shield the skin from UV damage, boost collagen production, and enhance skin elasticity. Cosmetic formulations incorporating grape stem extracts have displayed promising results in addressing various skin concerns, including reducing wrinkles, fine lines, and age spots, leading to a more youthful appearance. Additionally, grape stem extracts have exhibited anti-inflammatory properties, soothing irritated skin and diminishing redness. Exploring the potential of grape stem phenolic compounds for cosmetics paves the way for sustainable and natural beauty products. By harnessing the beauty benefits of grape stems, the cosmetics industry can provide effective and eco-friendly solutions for consumers seeking natural alternatives. Ongoing research holds the promise of innovative grape stem-based formulations that could revolutionize the cosmetics market, fully unlocking the potential of these extraordinary botanical treasures.
Collapse
Affiliation(s)
- Mónica Serra
- Mesosystem, Rua da Igreja Velha 295, São Félix da Marinha, 4410-160 Vila Nova de Gaia, Portugal
- CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
- LABBELS-Associate Laboratory, University of Minho, 4710-057 Braga, Portugal
| | - Ana Casas
- Mesosystem, Rua da Igreja Velha 295, São Félix da Marinha, 4410-160 Vila Nova de Gaia, Portugal
| | - José António Teixeira
- CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
- LABBELS-Associate Laboratory, University of Minho, 4710-057 Braga, Portugal
| | - Ana Novo Barros
- Mesosystem, Rua da Igreja Velha 295, São Félix da Marinha, 4410-160 Vila Nova de Gaia, Portugal
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
| |
Collapse
|
48
|
Sparks L, Whytock K, Divoux A, Sun Y, Pino M, Yu G, Smith S, Walsh M. A single nuclei atlas of aging human abdominal subcutaneous white adipose tissue. RESEARCH SQUARE 2023:rs.3.rs-3097605. [PMID: 37503028 PMCID: PMC10371078 DOI: 10.21203/rs.3.rs-3097605/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
White adipose tissue (WAT) is a robust energy storage and endocrine organ critical for maintaining metabolic health as we age. Our aim was to identify cell-specific transcriptional aberrations that occur in WAT with aging. We leveraged full-length snRNA-Seq to characterize the cellular landscape of human subcutaneous WAT in a prospective cohort of 10 Younger (≤ 30 years) and 10 Older individuals (≥ 65 years) balanced for sex and body mass index (BMI). We highlight that aging WAT is associated with adipocyte hypertrophy, increased proportions of resident macrophages (M2), an upregulated innate immune response and senescence profiles in specific adipocyte populations, highlighting CXCL14 as a biomarker of this process. We also identify novel markers of pre-adipocytes and track their expression levels through pre-adipocyte differentiation. We propose that aging WAT is associated with low-grade inflammation that is managed by a foundation of innate immunity to preserve the metabolic health of the WAT.
Collapse
Affiliation(s)
| | | | | | - Yifei Sun
- Icahn School of Medicine at Mount Sinai
| | - Maria Pino
- Translational Research Institute, AdventHealth
| | - Gongxin Yu
- Translational Research Institute, AdventHealth
| | | | | |
Collapse
|
49
|
Guzzi PH, Cortese F, Mannino GC, Pedace E, Succurro E, Andreozzi F, Veltri P. Analysis of age-dependent gene-expression in human tissues for studying diabetes comorbidities. Sci Rep 2023; 13:10372. [PMID: 37365269 DOI: 10.1038/s41598-023-37550-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023] Open
Abstract
The study of the relationship between type 2 diabetes mellitus (T2DM) disease and other pathologies (comorbidities), together with patient age variation, poses a challenge for medical research. There is evidence that patients affected by T2DM are more likely to develop comorbidities as they grow older. Variation of gene expression can be correlated to changes in T2DM comorbidities insurgence and progression. Understanding gene expression changes requires the analysis of large heterogeneous data at different scales as well as the integration of different data sources into network medicine models. Hence, we designed a framework to shed light on uncertainties related to age effects and comorbidity by integrating existing data sources with novel algorithms. The framework is based on integrating and analysing existing data sources under the hypothesis that changes in the basal expression of genes may be responsible for the higher prevalence of comorbidities in older patients. Using the proposed framework, we selected genes related to comorbidities from existing databases, and then analysed their expression with age at the tissues level. We found a set of genes that changes significantly in certain specific tissues over time. We also reconstructed the associated protein interaction networks and the related pathways for each tissue. Using this mechanistic framework, we detected interesting pathways related to T2DM whose genes change their expression with age. We also found many pathways related to insulin regulation and brain activities, which can be used to develop specific therapies. To the best of our knowledge, this is the first study that analyses such genes at the tissue level together with age variations.
Collapse
Affiliation(s)
- Pietro Hiram Guzzi
- Department of Surgical and Medical Sciences, Magna Graecia University, 88100, Catanzaro, Italy.
| | - Francesca Cortese
- Department of Surgical and Medical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Gaia Chiara Mannino
- Department of Surgical and Medical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Elisabetta Pedace
- Internal Medicine Unit, ASP Catanzaro, Soverato Hospital, Soverato, Italy
| | - Elena Succurro
- Department of Surgical and Medical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
- Internal Medicine Unit, R. Dulbecco Hospital, 88100, Catanzaro, Italy
| | - Francesco Andreozzi
- Department of Surgical and Medical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
- Internal Medicine Unit, R. Dulbecco Hospital, 88100, Catanzaro, Italy
| | | |
Collapse
|
50
|
Tarantini S, Subramanian M, Butcher JT, Yabluchanskiy A, Li X, Miller RA, Balasubramanian P. Revisiting adipose thermogenesis for delaying aging and age-related diseases: Opportunities and challenges. Ageing Res Rev 2023; 87:101912. [PMID: 36924940 PMCID: PMC10164698 DOI: 10.1016/j.arr.2023.101912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/03/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Adipose tissue undergoes significant changes in structure, composition, and function with age including altered adipokine secretion, decreased adipogenesis, altered immune cell profile and increased inflammation. Considering the role of adipose tissue in whole-body energy homeostasis, age-related dysfunction in adipose metabolism could potentially contribute to an increased risk for metabolic diseases and accelerate the onset of other age-related diseases. Increasing cellular energy expenditure in adipose tissue, also referred to as thermogenesis, has emerged as a promising strategy to improve adipose metabolism and treat obesity-related metabolic disorders. However, translating this strategy to the aged population comes with several challenges such as decreased thermogenic response and the paucity of safe pharmacological agents to activate thermogenesis. This mini-review aims to discuss the current body of knowledge on aging and thermogenesis and highlight the unexplored opportunities (cellular mechanisms and secreted factors) to target thermogenic mechanisms for delaying aging and age-related diseases. Finally, we also discuss the emerging role of thermogenic adipocytes in healthspan and lifespan extension.
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Madhan Subramanian
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Joshua T Butcher
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xinna Li
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Priya Balasubramanian
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|