1
|
Sun L, Wu Y, Yang J, Liang J, Li P, Yu X, Meng N, Sun T, Wang M, Chen C. The brain-white adipose tissue axis may play a crucial role in diabetes mellitus: a metabolic network analysis using total-body PET/CT imaging. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07337-5. [PMID: 40392299 DOI: 10.1007/s00259-025-07337-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
PURPOSE This study aims to construct an individualized glucose metabolism network using total-body 18F-FDG PET imaging, which provides a comprehensive view of glucose metabolism across various organs, to explore the role of inter-organ interactions in Diabetes Mellitus (DM). METHODS In this study, we constructed covariance metabolic networks using static total-body PET images, normalized by lean body mass, from 36 patients with DM (DM group) and 36 age- and sex-matched healthy controls (HC group). Differences in network properties between the DM and HC groups were evaluated at both group and individual levels. In addition, correlation analysis was performed to explore the relationship between network properties and baseline clinical data in the DM subjects. RESULTS We observed that the same edges in the first three edges with the largest values were brain-subcutaneous adipose tissue (SAT) and brain-visceral adipose tissue (VAT) at both group and individual levels. There was a positive correlation between the brain-VAT and BMI and there was a negative correlation between the brain-SAT and age. The most perturbed organ was the brain at both group and individual levels, and there was a positive correlation between the strength of abnormality of brain and age. CONCLUSION This study successfully used static total-body PET imaging to construct individualized glucose metabolism networks for patients with DM, identifying the brain-VAT and brain-SAT as the most significantly altered edge and the brain as the most affected organ. These findings provide novel insights into the role of the brain-white adipose tissue axis in glucose metabolism in DM.
Collapse
Affiliation(s)
- Lubing Sun
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Clinical Bioinformatics Experimental Center, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yaping Wu
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Junpeng Yang
- Department of Endocrinology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
| | - Junting Liang
- Clinical Bioinformatics Experimental Center, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
| | - Panlong Li
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Yu
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Meng
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Tao Sun
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Meiyun Wang
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Chuanliang Chen
- Department of Medical Imaging, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China.
- Clinical Bioinformatics Experimental Center, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, China.
| |
Collapse
|
2
|
Burdeau JA, Stephenson BJK, Chavarro JE, Mahalingaiah S, Preston EV, Hivert MF, Oken E, Calafat AM, Rifas-Shiman SL, Zota AR, James-Todd T. Early Pregnancy Plasma Per- and Polyfluoroalkyl Substances (PFAS) and Maternal Midlife Adiposity. J Clin Endocrinol Metab 2025; 110:e1966-e1974. [PMID: 39235224 DOI: 10.1210/clinem/dgae542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Indexed: 09/06/2024]
Abstract
CONTEXT Evidence suggests that exposure to per- and polyfluoroalkyl substances (PFAS) increases the risk of developing cardiometabolic disease risk factors. Limited research has evaluated associations between PFAS, assessed during pregnancy, a sensitive window for maternal endocrine effects, and long-term maternal adiposity. OBJECTIVE Estimate associations of early pregnancy measures of individual PFAS, and PFAS mixtures, with maternal adiposity in midlife. METHODS We studied 547 Project Viva participants with measures of early pregnancy (mean gestation 10.0 weeks; mean age 32.5 years) plasma concentrations of 6 PFAS and midlife adiposity outcomes (mean follow-up 17.7 years; mean age 50.7 years), including weight, waist circumference (WC), trunk fat mass (TFM), and total body fat mass (TBFM). We used linear regression and Bayesian Kernel Machine Regression (BKMR). RESULTS Linear regression estimated higher midlife weight per doubling of perfluorooctane sulfonate (PFOS) (3.8 kg [95% CI: 1.6, 5.9]) and 2-(N-ethyl-perfluorooctane sulfonamido) acetate (2.3 kg [95% CI: 0.9, 3.7]). BKMR analyses of single PFAS plasma concentrations (comparing the 25th percentile concentration to the 75th percentile) showed a positive association between PFOS and midlife adiposity (weight: 7.7 kg [95% CI: 4.0, 11.5]; TFM: 1.2 kg [95% CI: 0.0, 2.3]; TBFM: 3.0 kg [95% CI: 0.8, 5.2]), but inverse associations with perfluorononanoate (weight: -6.0 kg [95% CI: -8.5, -3.5]; WC: -1.8 cm [95% CI: -3.2, -0.3]; TFM: -0.8 kg [95% CI: -1.5, -0.1]; TBFM: -1.4 kg [95% CI: -2.7, -0.3]) and perfluorohexane sulfonate (TFM: -0.8 kg [95% CI: -1.5, -0.1]; TBFM: -1.4 kg [95% CI: -2.6, -0.2]). No associations were observed with the overall PFAS mixture. CONCLUSION Select PFAS, assessed in pregnancy, may differentially affect maternal midlife adiposity, influencing later-life maternal cardiometabolic health.
Collapse
Affiliation(s)
- Jordan A Burdeau
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Briana J K Stephenson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Jorge E Chavarro
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Shruthi Mahalingaiah
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emma V Preston
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emily Oken
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA
| | - Ami R Zota
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Tamarra James-Todd
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| |
Collapse
|
3
|
Chen C, Wang L. Aging and metabolic dysfunction-associated steatotic liver disease: a bidirectional relationship. Front Med 2025:10.1007/s11684-025-1133-7. [PMID: 40316793 DOI: 10.1007/s11684-025-1133-7] [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: 10/28/2024] [Accepted: 01/09/2025] [Indexed: 05/04/2025]
Abstract
In recent years, aging and cellular senescence have triggered an increased interest in corresponding research fields. Evidence shows that the complex aging process is involved in the development of many chronic liver diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). In fact, aging has a tremendous effect on the liver, leading to a gradual decline in the metabolism, detoxification and immune functions of the liver, which in turn increases the risk of liver disease. These changes can be based on the aging of liver cells (hepatocytes, liver sinusoidal endothelial cells, hepatic stellate cells, and Kupffer cells). Similarly, patients with liver diseases exhibit increases in the aging phenotype and aging cells, often manifesting as faster physical functional decline, which is closely related to the promoting effect of liver disease on aging. This review summarizes the interplay between MASLD/MASH development and aging, aiming to reveal the complex relationships that exacerbate one another. Moreover, the corresponding schemes for delaying aging or treating diseases are discussed to provide a basis for the development of effective prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
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
|
5
|
Borges GA, Diaz-delCastillo M, Guilatco AJ, El-Masri BM, Mustapha FA, Gundesen MT, Hinge M, Lund T, Abdallah N, Baughn LB, Xu M, Gingery A, Tchkonia T, Kirkland JL, Kourelis T, Drake MT, Andersen TL, Weivoda MM. Senescence profiling of monoclonal gammopathies reveals paracrine senescence as a crucial defense against disease progression. Leukemia 2025; 39:1206-1217. [PMID: 40164720 PMCID: PMC12055601 DOI: 10.1038/s41375-025-02572-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
Multiple myeloma (MM) is a plasma cell (PC) malignancy that is preceded by monoclonal gammopathy of undetermined significance (MGUS) and/or smoldering multiple myeloma (SMM). MGUS and SMM PCs exhibit the same primary oncogenic abnormalities as MM but lack the end-organ damage that defines proliferative disease, suggesting that clonal PCs in these precursor conditions could exhibit senescence or senescence-like growth arrest. Herein we identified monoclonal gammopathy patient-derived PCs that exhibit senescence features and found that senescent PCs were significantly increased in MGUS patients compared to SMM or MM. Spatial analysis of senescent PCs in stable MGUS and SMM patient biopsies demonstrated the activation of local paracrine senescence in the bone marrow microenvironment. Stable MGUS and SMM patients also exhibited disease-specific senescence-associated secretory phenotype (SASP) signatures that significantly correlated with PC burden and clonal antibody. In contrast, progressing MGUS, SMM, and new MM patients lacked local paracrine senescence responses and robust activation of disease specific SASP signatures. Overall, these data suggest that failure to activate tumor-specific paracrine senescence responses is key to disease progression in monoclonal gammopathies.
Collapse
Affiliation(s)
| | - Marta Diaz-delCastillo
- University of Aarhus, Department of Forensic Medicine, Aarhus, Denmark
- Danish Spatial Imaging Consortium (DanSIC), University of Aarhus and University of Southern Denmark, Aarhus, Denmark
| | - Angelo J Guilatco
- Mayo Clinic Division of Hematology, Rochester, MN, USA
- University of Michigan, Cancer Biology Graduate Program, Ann Arbor, MI, USA
| | | | - Fatima A Mustapha
- University of Aarhus, Department of Forensic Medicine, Aarhus, Denmark
| | | | - Maja Hinge
- Lillebaelt Hospital, Department of Hematology, Vejle, Denmark
| | - Thomas Lund
- Odense University Hospital, Department of Hematology, Odense, Denmark
| | | | - Linda B Baughn
- Mayo Clinic Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Ming Xu
- University of Minnesota Department of Biochemistry, Molecular Biology and Biophysics, Minneapolis, MN, USA
| | - Anne Gingery
- Mayo Clinic Department of Orthopedic Surgery, Rochester, MN, USA
- Mayo Clinic Department of Biochemistry and Molecular Biology, Rochester, MN, USA
| | - Tamar Tchkonia
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Matthew T Drake
- Hospital for Special Surgery Division of Endocrinology, New York, NY, USA
| | - Thomas Levin Andersen
- University of Aarhus, Department of Forensic Medicine, Aarhus, Denmark.
- Danish Spatial Imaging Consortium (DanSIC), University of Aarhus and University of Southern Denmark, Aarhus, Denmark.
- University of Southern Denmark, Odense, Denmark.
- Odense University Hospital, Department of Clinical Pathology, Odense, Denmark.
| | - Megan M Weivoda
- Mayo Clinic Division of Hematology, Rochester, MN, USA.
- Mayo Clinic Department of Biochemistry and Molecular Biology, Rochester, MN, USA.
| |
Collapse
|
6
|
Nishimura ES, Hishikawa A, Nakamichi R, Akashio R, Chikuma S, Hashiguchi A, Yoshimoto N, Hama EY, Maruki T, Itoh W, Yamaguchi S, Yoshino J, Itoh H, Hayashi K. DNA damage in proximal tubules triggers systemic metabolic dysfunction through epigenetically altered macrophages. Nat Commun 2025; 16:3958. [PMID: 40295524 PMCID: PMC12037803 DOI: 10.1038/s41467-025-59297-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
DNA damage repair is a critical physiological process closely linked to aging. The accumulation of DNA damage in renal proximal tubular epithelial cells (PTEC) is related to a decline in kidney function. Here, we report that DNA double-strand breaks in PTECs lead to systemic metabolic dysfunction, including weight loss, reduced fat mass, impaired glucose tolerance with mitochondrial dysfunction, and increased inflammation in adipose tissues and the liver. Single-cell RNA sequencing analysis reveals expansion of CD11c+ Ccr2+ macrophages in the kidney cortex, liver, and adipose tissues and Ly6Chi monocytes in peripheral blood. DNA damage in PTECs is associated with hypomethylation of macrophage activation genes, including Gasdermin D, in peripheral blood cells, which is linked to reduced DNA methylation at KLF9-binding motifs. Macrophage depletion ameliorates metabolic abnormalities. These findings highlight the impact of kidney DNA damage on systemic metabolic homeostasis, revealing a kidney-blood-metabolism axis mediated by epigenetic changes in macrophages.
Collapse
Affiliation(s)
- Erina Sugita Nishimura
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihito Hishikawa
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ran Nakamichi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Riki Akashio
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shunsuke Chikuma
- Department of Immunology, Keio University School of Medicine, Tokyo, Japan
- College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu City, Taiwan
| | - Akinori Hashiguchi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Norifumi Yoshimoto
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Eriko Yoshida Hama
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tomomi Maruki
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Wataru Itoh
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Yamaguchi
- School of Medicine Medical Education Center, Keio University, Tokyo, Japan
| | - Jun Yoshino
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan
- The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Japan
| | - Hiroshi Itoh
- Center for Preventive Medicine, Keio University Hospital, Tokyo, Japan
| | - Kaori Hayashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| |
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
|
Zhu T, Tian Y, Wang J, Wu Z, Xie W, Liu H, Li X, Tao L, Guo X. The Relationship between Visceral Fat Accumulation and Risk of Cardiometabolic Multimorbidity: The Roles of Accelerated Biological Aging. Nutrients 2025; 17:1397. [PMID: 40284259 PMCID: PMC12030224 DOI: 10.3390/nu17081397] [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: 03/20/2025] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
OBJECTIVES To investigate the association between visceral fat accumulation and the risk of cardiometabolic multimorbidity (CMM) and the potential roles of accelerated biological aging in this relationship. METHODS Using data from the UK Biobank, a nationwide cohort study was conducted using the available baseline body roundness index (BRI) measurement. Biological aging was assessed using the Klemera-Doubal method for biological age and the phenotypic age algorithms. The association between the BRI and CMM was estimated using the Cox proportional hazards regression model, while the roles of biological aging were examined through interaction and mediation analyses. RESULTS During a median follow-up of 14.52 years, 6156 cases of CMM were identified. A significant association was observed between the BRI and CMM. The hazard ratio (HR) for CMM was 3.72 (95% confidence interval [CI]: 3.35-4.13) for individuals in the highest quartile compared with those in the lowest quartile of the BRI. More importantly, the BRI (AUC, 0.701; 95% CI, 0.694-0.707) demonstrated superior predictive performance relative to body mass index (AUC, 0.657; 95% CI, 0.650-0.664). Furthermore, the BRI exhibited additive interactions with accelerated biological aging on the risk of CMM, and accelerated biological aging partially mediated the association between the BRI and CMM. CONCLUSIONS These findings provide evidence for the application of the BRI as a novel and readily accessible screening tool associated with CMM, suggesting that the effective management of visceral fat and biological aging deceleration may hold promise for reducing CMM risk.
Collapse
Affiliation(s)
- Tianyu Zhu
- Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
| | - Yixing Tian
- Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
| | - Jinqi Wang
- Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
| | - Zhiyuan Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Wenhan Xie
- Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
| | - Haotian Liu
- Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Melbourne, VIC 3086, Australia
| | - Lixin Tao
- Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
| | - Xiuhua Guo
- Beijing Key Laboratory of Environment and Aging, School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia
- Department of Epidemiology and Health Statistics, School of the Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmen Street, Beijing 100069, China
| |
Collapse
|
9
|
Altea-Manzano P, Decker-Farrell A, Janowitz T, Erez A. Metabolic interplays between the tumour and the host shape the tumour macroenvironment. Nat Rev Cancer 2025; 25:274-292. [PMID: 39833533 DOI: 10.1038/s41568-024-00786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Metabolic reprogramming of cancer cells and the tumour microenvironment are pivotal characteristics of cancers, and studying these processes offer insights and avenues for cancer diagnostics and therapeutics. Recent advancements have underscored the impact of host systemic features, termed macroenvironment, on facilitating cancer progression. During tumorigenesis, these inherent features of the host, such as germline genetics, immune profile and the metabolic status, influence how the body responds to cancer. In parallel, as cancer grows, it induces systemic effects beyond the primary tumour site and affects the macroenvironment, for example, through inflammation, the metabolic end-stage syndrome of cachexia, and metabolic dysregulation. Therefore, understanding the intricate metabolic interplay between the tumour and the host is a growing frontier in advancing cancer diagnosis and therapy. In this Review, we explore the specific contribution of the metabolic fitness of the host to cancer initiation, progression and response to therapy. We then delineate the complex metabolic crosstalk between the tumour, the microenvironment and the host, which promotes disease progression to metastasis and cachexia. The metabolic relationships among the host, cancer pathogenesis and the consequent responsive systemic manifestations during cancer progression provide new perspectives for mechanistic cancer therapy and improved management of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Ayelet Erez
- Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
10
|
Khalafi M, Kheradmand S, Habibi Maleki A, Symonds ME, Rosenkranz SK, Batrakoulis A. The Effects of Concurrent Training Versus Aerobic or Resistance Training Alone on Body Composition in Middle-Aged and Older Adults: A Systematic Review and Meta-Analysis. Healthcare (Basel) 2025; 13:776. [PMID: 40218073 PMCID: PMC11989159 DOI: 10.3390/healthcare13070776] [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: 02/02/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Introduction and Aim: The beneficial effects of aerobic training (AT) on preventing excess fat mass, and of resistance training (RT) on skeletal muscle adaptation, are well established. However, the effects of concurrent training (CT) compared to AT or RT alone on body composition in middle-aged and older adults are less understood, and therefore, the focus of this meta-analysis. Methods: Three databases, including PubMed, Web of Science, and Scopus, were searched from inception to March 2024. Randomized trials were included if they compared CT versus either AT or RT, and included body composition measures such as fat mass, body fat percentage, waist circumference, visceral fat mass, lean body mass (LBM), muscle mass/volume, or muscle or muscle fiber cross-sectional area (CSA), in middle-aged (50 to <65 years) and older adults (≥65 years). Weighted mean differences (WMD) or standardized mean differences (SMD) and 95% confidence intervals (CIs) were calculated using random effects models. Results: A total of 53 studies involving 2873 participants were included. Overall, CT increased body weight and LBM significantly more, trending toward significantly larger increases in muscle mass and CSA, compared with AT alone. However, there were no significant differences between CT and RT alone, for body weight, BMI, body fat percentage, fat mass, waist circumference, or visceral fat mass. Conclusions: CT is as effective as AT for decreasing body fat measures and as effective as RT for increasing muscle mass in middle-aged and older adults, and it should be recommended accordingly.
Collapse
Affiliation(s)
- Mousa Khalafi
- Department of Sport Sciences, Faculty of Humanities, University of Kashan, Kashan 87317-53153, Iran
| | - Shokoufeh Kheradmand
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Mazandaran, Babolsar 47416-13534, Iran;
| | - Aref Habibi Maleki
- Physiology Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran;
| | - Michael E. Symonds
- Centre for Perinatal Research, Academic Unit of Population and Lifespan Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK;
| | - Sara K. Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | - Alexios Batrakoulis
- Department of Physical Education and Sport Science, Democritus University of Thrace, 69100 Komotini, Greece
- Department of Physical Education and Sport Science, University of Thessaly, 42100 Trikala, Greece
| |
Collapse
|
11
|
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
|
12
|
Conover CA. Cellular senescence and PAPP-A. Growth Horm IGF Res 2025; 80:101637. [PMID: 39904113 DOI: 10.1016/j.ghir.2025.101637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Cellular senescence and its accompanying secretome have major impact on growth and aging of mammalian organisms. A novel protease, PAPP-A, regulates the bioavailability of an important growth factor, insulin-like growth factor (IGF)-I, and has major impact on growth and aging. This short review will explore a new perspective of cellular senescence and PAPP-A.
Collapse
Affiliation(s)
- Cheryl A Conover
- Department of Endocrinology, Endocrine Research Unit, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States of America.
| |
Collapse
|
13
|
Chen C, Tang P, Zhu W. Systemic immune-inflammation index mediates the association between abdominal obesity and serum klotho levels. Sci Rep 2025; 15:4205. [PMID: 39905076 PMCID: PMC11794886 DOI: 10.1038/s41598-025-88015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
The weight-adjusted waist index (WWI) has emerged as a reliable indicator of abdominal obesity. α-Klotho, a transmembrane protein, functions as a suppressor of aging. However, the relationship between these two factors remains underexplored. This study aims to investigate the association between WWI and serum α-Klotho levels in middle-aged and elderly Americans, with a focus on exploring the potential mediating role of the systemic immune inflammation index (SII). A cross-sectional study was conducted using data from 6997 middle-aged and elderly Americans participating in the National Health and Nutrition Examination Surveys (NHANES) between 2011 and 2016. Multiple linear regression analysis was employed to assess the relationship between WWI and serum α-Klotho concentrations. Additionally, mediation analysis was performed to investigate the mediating effect of SII on the relationships. Our analysis revealed a significant negative correlation between WWI and serum α-Klotho levels in the survey-weighted multiple linear regression models (adjusted percent change: -7.79; 95% CI: -10.15, -5.37). Mediation analysis demonstrated that the association between WWI and α-Klotho levels was partially mediated by SII (adjusted percent change: -0.88; 95% CI: -1.24, -0.45), with the proportion of mediation amounting to 11.6%. Further age-stratified results showed that the mediating role of SII was more pronounced among individuals aged ≥ 60 years, exhibiting a mediating effect of 26.3%, in contrast to 4.2% for those < 60 years. The findings suggest that WWI is inversely associated with serum α-Klotho concentrations and that this association is partially mediated by SII, especially in older people.
Collapse
Affiliation(s)
- Chenchun Chen
- Institute of Public Health, Guangzhou Medical University & Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Peng Tang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, China
| | - Wei Zhu
- Institute of Public Health, Guangzhou Medical University & Guangzhou Center for Disease Control and Prevention, Guangzhou, China.
- Institute of Public Health, Guangzhou Medical University & Department of Toxicology, Guangzhou Center for Disease Control and Prevention, No. 1, Qide Road, Jiahe, Baiyun District, Guangzhou, 510440, China.
| |
Collapse
|
14
|
Romero M, Gelsomini A, Miller K, Suresh D, Thaller S, Frasca D. In Vitro Treatment with Metformin Significantly Reduces Senescent B Cells Present in the Adipose Tissue of People with Obesity. J Nutr 2025; 155:445-452. [PMID: 39389182 PMCID: PMC11867121 DOI: 10.1016/j.tjnut.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Our previous work has shown that senescent B cells accumulate in the human adipose tissue (AT) of people with obesity, where they express transcripts for markers associated with the senescence-associated secretory phenotype (SASP) and secrete multiple inflammatory mediators. These functions of AT-derived B cells are metabolically supported. OBJECTIVES To show that Metformin (MET), a widely used hypoglycemic and antidiabetic drug, is able at least in vitro to decrease frequencies, secretory profile, and metabolic requirements of senescent B cells isolated from the AT of people with obesity. METHODS We recruited adult females with obesity (n = 8, age 40 ± 2 y, BMI range: 33-42) undergoing breast reduction surgery, who donated their discarded subcutaneous AT. B cells from stromal vascular fractions isolated after collagenase digestion of the AT were evaluated after in vitro incubation with MET (1 mM × 106 B cells) or with a control medium without MET for the following measures: expression of transcripts for SASP-associated markers (p16INK4a and p21CIP1/WAF1) measured by quantitative polymerase chain reaction (qPCR); secretion of inflammatory cytokines (TNF-α, IL-6, IFN-γ and IL-17A) measured by a Cytometric Bead Array); metabolic characteristics as identified by a glycolytic test and Seahorse technology, and by the expression of transcripts for glucose transporters and metabolic enzymes involved in glucose metabolic pathways, measured by qPCR. To examine differences between MET-treated compared with untreated groups, paired Student's t tests (two-tailed) were employed. RESULTS MET in vitro was able to reduce frequencies and numbers of senescent B cells, as identified by staining with β-galactosidase, as well as the secretion of inflammatory cytokines, the expression of transcripts for SASP, and metabolic markers that support intrinsic B cell inflammation. CONCLUSIONS Our results provide evidence to support the beneficial effects of MET in reducing AT-related inflammation through its effects on senescent B cells.
Collapse
Affiliation(s)
- Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Andrew Gelsomini
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kate Miller
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Dhananjay Suresh
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Seth Thaller
- DeWitt Daughtry Family Department of Surgery, Division of Plastic and Reconstructive Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States.
| |
Collapse
|
15
|
Vieira FT, Cai Y, Gonzalez MC, Goodpaster BH, Prado CM, Haqq AM. Poor muscle quality: A hidden and detrimental health condition in obesity. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09941-0. [PMID: 39833502 DOI: 10.1007/s11154-025-09941-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Poor muscle quality (MQ) is a hidden health condition in obesity, commonly disregarded and underdiagnosed, associated with poor health-related outcomes. This narrative review provides an in-depth exploration of MQ in obesity, including definitions, available assessment methods and challenges, pathophysiology, association with health outcomes, and potential interventions. MQ is a broad term that can include imaging, histological, functional, or metabolic assessments, evaluating beyond muscle quantity. MQ assessment is highly heterogeneous and requires further standardization. Common definitions of MQ include 1) muscle-specific strength (or functional MQ), the ratio between muscle strength and muscle quantity, and 2) muscle composition (or morphological MQ), mainly evaluating muscle fat infiltration. An individual with obesity might still have normal or higher muscle quantity despite having poor MQ, and techniques for direct measurements are needed. However, the use of body composition and physical function assessments is still limited in clinical practice. Thus, more accessible techniques for assessing strength, muscle mass, and composition should be further explored. Obesity leads to adipocyte dysfunction, generating a low-grade chronic inflammatory state, which leads to mitochondrial dysfunction. Adipocyte and mitochondrial dysfunction result in metabolic dysfunction manifesting clinically as insulin resistance, dyslipidemia, and fat infiltration into organs such as muscle, which in excess is termed myosteatosis. Myosteatosis decreases muscle cell function and insulin sensitivity, creating a vicious cycle of inflammation and metabolic derangements. Myosteatosis increases the risk of poor muscle function, systemic metabolic complications, and mortality, presenting prognostic potential. Interventions shown to improve MQ include nutrition, physical activity/exercise, pharmacology, and metabolic and bariatric surgery.
Collapse
Affiliation(s)
- Flavio T Vieira
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Yuanjun Cai
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - M Cristina Gonzalez
- Postgraduate Program in Nutrition and Food, Federal University of Pelotas, Pelotas, Rio Grande Do Sul, Brazil
| | | | - Carla M Prado
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada.
| | - Andrea M Haqq
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
16
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
17
|
Onat E, Kocaman N, Hançer S, Yildirim M. Role of Dardarin and Isthmin-1 in the Protective Effect of Hydroxytyrosol Against Corn Syrup-Induced Liver Damage. Cureus 2025; 17:e76803. [PMID: 39897276 PMCID: PMC11787043 DOI: 10.7759/cureus.76803] [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] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
INTRODUCTION In this study, it was investigated whether dardarin (LRRK2) and isthmin-1 (ISM1) play a role in the protective effect of hydroxytyrosol (HT) used to prevent liver damage caused by corn syrup in rats. METHODS Rats were divided into four groups with six in each group: 1) control, 2) HT, 3) corn syrup, and 4) corn syrup + HT. Rats were given water containing 30% corn syrup for six weeks. At the same time, HT-containing liquid was given orally at 4 ml/kg/day, alone and together with corn syrup for six weeks. The weights of the rats were measured every week. LRRK2 and ISM1 molecules in liver tissue were evaluated by histopathological methods. Biochemical parameters were also examined with the enzyme-linked immunosorbent assay (ELISA) method. RESULTS It was found that weight gain was less in rats receiving HT than in those consuming corn syrup. The increase in cholesterol, triglyceride, and liver enzyme levels because of corn syrup consumption decreased with HT consumption. As a result of histopathological analysis, it was observed that the increase in LRRK2 and ISM1 levels observed in the liver tissue in the corn syrup-administered group decreased when HT was administered together with corn syrup. In addition, it was determined that the increase in sinusoidal expansion and hepatocyte necrosis observed in the liver tissue as a result of corn syrup application decreased as a result of the application of HT together with corn syrup. CONCLUSION The protective effect of HT against the damage caused by corn syrup in the liver has been demonstrated once again; however, LRRK2 and ISM1 are thought to contribute to this issue.
Collapse
Affiliation(s)
- Elif Onat
- Department of Medical Pharmacology, Fırat University, Elazığ, TUR
| | - Nevin Kocaman
- Department of Histology and Embryology, Fırat University, Elazığ, TUR
| | - Serhat Hançer
- Department of Histology and Embryology, Fırat University, Elazığ, TUR
| | - Murat Yildirim
- Department of Pediatrics, University of Wisconsin-Madison, Madison, USA
| |
Collapse
|
18
|
Locke B, Lu R. Establishment of immortalized porcine intramuscular preadipocytes for the study of lipid metabolism. Biochem Cell Biol 2025; 103:1-11. [PMID: 40127467 DOI: 10.1139/bcb-2024-0174] [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: 03/26/2025] Open
Abstract
Intramuscular adipose tissue is associated with an increased risk for the development of metabolic syndrome. A cellular model of adipogenesis in muscular tissues would be an invaluable tool for studying regulatory factors in this important process. Cellular stress can impact the homeostasis of various metabolic pathways, including lipid metabolism. In this study, a porcine intramuscular preadipocyte cell line was established, which displayed mature adipocyte attributes such as lipid accumulation and increased expression of adipogenic gene markers. Since it is well established that endoplasmic reticulum (ER) and Golgi stress impact adipogenesis, we sought to investigate the effects of ER/Golgi stress and an associated protein, CREB3, in this cell line model. We found that this novel model maintains robust adipogenic capabilities, and that ER stress can negatively affect adipogenic markers. Overall, these findings demonstrate the strength of the new cell model for studying adipogenesis, and highlight the impact of ER stress on lipid metabolism.
Collapse
Affiliation(s)
- Briana Locke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
19
|
Su Z, Xu T, Sun JY, Sun W, Kong X. Alterations in the transcriptome and microRNAs of adipose-derived mesenchymal stem cells from different sites in rats during aging. Am J Physiol Cell Physiol 2025; 328:C78-C94. [PMID: 39495250 DOI: 10.1152/ajpcell.00044.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Aging is an intricate and gradual process characterized by tissue and cellular dysfunction. Adipose-derived mesenchymal stem cells (ADMSCs) experience a functional decline as part of systemic aging. However, the alterations in ADMSCs across various anatomical sites throughout an individual's lifespan remain unclear. To shed light on these changes, we collected white adipose tissue and brown adipose tissue samples from the epididymis, perirenal, inguinal, and scapular regions of young, adult, and aged rats and subsequently isolated ADMSCs for RNA sequencing. As aging progressed, we observed a reduction in the number of ADMSCs at all anatomical sites. Marker genes of ADMSCs from different sites were identified. Aging triggered notable activation of inflammatory and immune responses while diminishing the ADMSC differentiation capacity and ability to maintain a normal tissue morphology. Furthermore, miR-195-5p and miR-497-3p, which promoted cell senescence and apoptosis while inhibiting proliferation and differentiation, were positively correlated with aging. These findings increase our understanding of ADMSC senescence and underscore the unique physiological changes and functions of ADMSCs across different anatomical sites during aging.NEW & NOTEWORTHY Dynamic changes in mRNAs and miRNAs of ADMSCs during aging are shown. As aging progressed, we observed a reduction in the number of ADMSCs at all anatomical sites. Aging leads to the activation of inflammatory and cellular dysfunction. miR-195-5p and miR-497-3p are positively correlated with aging, which promoted cell senescence and apoptosis while inhibiting proliferation and differentiation. ADMSCs associated with different anatomical sites have site-specific markers.
Collapse
Affiliation(s)
- Zhenyang Su
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, People's Republic of China
| | - Tianhua Xu
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, People's Republic of China
| | - Jin-Yu Sun
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, People's Republic of China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
20
|
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
|
21
|
Lee Y, Tassey J, Sarkar A, Levi JN, Lee S, Liu NQ, Drake AC, Nguyen F, Magallanes J, Stevic U, Lu J, Ge D, Tang H, Mkaratigwa T, Yang J, Bian F, Shkhyan R, Bonaguidi MA, Evseenko D. Pharmacological inactivation of a non-canonical gp130 signaling arm attenuates chronic systemic inflammation and multimorbidity induced by a high-fat diet. Sci Rep 2024; 14:31151. [PMID: 39732741 PMCID: PMC11682372 DOI: 10.1038/s41598-024-82414-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Interleukin-6 (IL-6) is a major pro-inflammatory cytokine that demonstrates a robust correlation with age and body mass index (BMI) as part of the senescence-associated secretory phenotype. IL-6 cytokines also play a crucial role in metabolic homeostasis and regenerative processes primarily via the canonical STAT3 pathway. Thus, selective modulation of IL-6 signaling may offer a unique opportunity for therapeutic interventions. Our recent studies identified a novel non-canonical signaling pathway that involves prolonged activation of SRC family of kinases (SFKs) by IL-6/gp130, where genetic or pharmacological inhibition of this pathway was protective in several acute injury models. This study was designed to assess the effect of a small molecule (R159) that inhibits the non-canonical signaling in a mouse model of multimorbidity induced by chronic inflammation. Aged mice were fed a high-fat diet (HFD) to exacerbate chronic inflammation and inflammaging-related conditions, and R159 significantly decreased systemic inflammatory responses in adipose tissue and liver. R159 was protective against trabecular bone and articular cartilage loss and markedly prevented neurogenesis decline. Moreover, R159 reduced weight gain induced by HFD and increased physical activity levels. These findings suggest that selective pharmacological inhibition of SFK signaling downstream of IL6/gp130 offers a promising strategy to alleviate systemic chronic inflammation and relevant multimorbidity.
Collapse
Affiliation(s)
- Youngjoo Lee
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Jade Tassey
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arijita Sarkar
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jonathan N Levi
- Department of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Siyoung Lee
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Nancy Q Liu
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew C Drake
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Falisha Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Jenny Magallanes
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Una Stevic
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Jinxiu Lu
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dawei Ge
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Orthopedics Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hanhan Tang
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Tadiwanashe Mkaratigwa
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jichen Yang
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Fangzhou Bian
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ruzanna Shkhyan
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael A Bonaguidi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, USC, Los Angeles, CA, USA
- Department of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, USC, Los Angeles, CA, USA.
| |
Collapse
|
22
|
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
|
23
|
Kartika RW, Timotius KH, Sidharta VM, Djuartina T, Sartika CR. Aging Parameters of the Accelerated Aging Procedure through D-Galactose Induction. ACTA MEDICA PHILIPPINA 2024; 58:104-109. [PMID: 39830419 PMCID: PMC11739527 DOI: 10.47895/amp.vi0.7801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background and Objectives Intraperitoneal injection (i.p.) of D-galactose (D-gal) accelerates aging and develops aging models. A low dose of long-term use and a high dose of short-term use of D-gal can induce natural aging in mice, like brain, cardiac, liver, renal, and skin aging, and erectile dysfunction. Our research aims to determine whether a high dose of short-term use of D-gal. i.p. in rats can induce natural aging and affect the following parameters: body weight (BW), Superoxide Dismutase (SOD), Vascular endothelial growth factor (VEGF), C-reactive protein (CRP), and myostatin. Methods A daily D-gal i.p. dose of 300 mg/ml/kg for seven days was carried out to induce aging parameters in the rats. After seven days, the body and gastrocnemius circumference of the rats were weighed, and biochemical analysis for SOD, VEGF, CRP, and myostatin in the blood plasma was done. Results The data obtained were analyzed using nonparametric statistics Friedman test and Mann-Whitney test. After the seven day-intervention, both the control (NaCl 0.9% i.p.) and the high dose of short-term use of D-gal i.p. groups showed no significant difference in the body weight and gastrocnemius circumference. However, D-gal administration could increase the blood plasma level of SOD, VEGF, CRP, and myostatin. Conclusion We conclude that a high dose of short-term intraperitoneal D-galactose can be administrated to induce aging in rat models. The SOD, VEGF, CRP and myostatin can be used as aging parameters.
Collapse
Affiliation(s)
- Ronald Winardi Kartika
- Faculty of Medicine and Health Science, Krida Wacana Christian University, Jakarta, Indonesia
| | - Kris Herawan Timotius
- Faculty of Medicine and Health Science, Krida Wacana Christian University, Jakarta, Indonesia
| | | | - Tena Djuartina
- School of Medicine Universitas Katolik Indonesia Atma Jaya, Jakarta, Indonesia
| | | |
Collapse
|
24
|
Elmitwalli O, Darwish R, Al-Jabery L, Algahiny A, Roy S, Butler AE, Hasan AS. The Emerging Role of p21 in Diabetes and Related Metabolic Disorders. Int J Mol Sci 2024; 25:13209. [PMID: 39684919 DOI: 10.3390/ijms252313209] [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: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
In the context of cell cycle inhibition, anti-proliferation, and the dysregulation observed in certain cancer pathologies, the protein p21 assumes a pivotal role. p21 links DNA damage responses to cellular processes such as apoptosis, senescence, and cell cycle arrest, primarily functioning as a regulator of the cell cycle. However, accumulating empirical evidence suggests that p21 is both directly and indirectly linked to a number of different metabolic processes. Intriguingly, recent investigations indicate that p21 significantly contributes to the pathogenesis of diabetes. In this review, we present a comprehensive evaluation of the scientific literature regarding the involvement of p21 in metabolic processes, diabetes etiology, pancreatic function, glucose homeostasis, and insulin resistance. Furthermore, we provide an encapsulated overview of therapies that target p21 to alleviate metabolic disorders. A deeper understanding of the complex interrelationship between p21 and diabetes holds promise for informing current and future therapeutic strategies to address this rapidly escalating health crisis.
Collapse
Affiliation(s)
- Omar Elmitwalli
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Radwan Darwish
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Lana Al-Jabery
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ahmed Algahiny
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Sornali Roy
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Alexandra E Butler
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ammar S Hasan
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| |
Collapse
|
25
|
Salinero AE, Venkataganesh H, Abi-Ghanem C, Riccio D, Kelly RD, Gannon OJ, Sura A, Brooks HL, Zuloaga DG, Zuloaga KL. Effects of high fat diet on metabolic health vary by age of menopause onset. Int J Obes (Lond) 2024; 48:1839-1843. [PMID: 39152337 PMCID: PMC11585410 DOI: 10.1038/s41366-024-01618-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Menopause accelerates metabolic dysfunction, including (pre-)diabetes, obesity and visceral adiposity. However, the effects of endocrine vs. chronological aging in this progression are poorly understood. We hypothesized that menopause, especially in the context of middle-age, would exacerbate the metabolic effects of a high fat diet. Using young-adult and middle-aged C57BL/6J female mice, we modeled diet-induced obesity via chronic administration of high fat (HF) diet vs. control diet. We modeled peri-menopause/menopause via injections of 4-vinylcyclohexene diepoxide, which accelerates ovarian failure vs. vehicle. We performed glucose tolerance tests 2.5 and 7 months after diet onset, during the peri-menopausal and menopausal phases, respectively. Peri-menopause increased the severity of glucose intolerance and weight gain in middle-aged, HF-fed mice. Menopause increased weight gain in all mice regardless of age and diet, while chronological aging drove changes in adipose tissue distribution towards more visceral vs. subcutaneous adiposity. These data are in line with clinical data showing that post-menopausal women are more susceptible to metabolic dysfunction and suggest that greater chronological age exacerbates the effects of endocrine aging (menopause). This work highlights the importance of considering both chronological and endocrine aging in studies of metabolic health.
Collapse
Affiliation(s)
- Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Riccio
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Avi Sura
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Heddwen L Brooks
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Damian G Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY, 12222, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
26
|
Park JY, Chung YJ, Song JY, Kil KC, Lee HY, Chae J, Kim MR. Sarcopenic Obesity: A Comprehensive Approach for Postmenopausal Women. J Menopausal Med 2024; 30:143-151. [PMID: 39829191 PMCID: PMC11745730 DOI: 10.6118/jmm.24004] [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: 03/07/2024] [Revised: 09/07/2024] [Accepted: 10/04/2024] [Indexed: 01/22/2025] Open
Abstract
Sarcopenic obesity, characterized by the concurrent presence of muscle loss and obesity, poses significant health challenges, especially in the elderly. This review explores the impact of sarcopenic obesity on disability, metabolic health, comorbidities, and potential management strategies. With the aging global population, the prevalence of sarcopenic obesity is expected to increase, necessitating a comprehensive management approach. Early screening, prevention, and ongoing research on its underlying mechanisms and therapeutic options are crucial for promoting healthy aging.
Collapse
Affiliation(s)
- Jung Yoon Park
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Youn-Jee Chung
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae-Yen Song
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki Cheol Kil
- Department of Obstetrics and Gynecology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hong Yeon Lee
- Department of Obstetrics and Gynecology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jungwon Chae
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mee-Ran Kim
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
27
|
Liu C, He E, Fu P, He L, Zhou L, Lou A, Liu Y, Fu H, Shen Q, Luo J, Quan W. Quality Characteristics Changes of the Fat Portion of Chinese Bacon During Processing Based on Physicochemical Properties and Microstructure Studies. Foods 2024; 13:3821. [PMID: 39682893 DOI: 10.3390/foods13233821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
In order to elucidate the development of quality properties in the fat portion of Chinese bacon during low-temperature smoking (LTS), raw pork was cured for five days, followed by infusion with smoked liquid and a subsequent ten-day smoking period characterized by alternating high and low-temperature conditions. The physicochemical characteristics and microstructures of the fat portion of the Chinese bacon were examined at three stages: the raw meat stage (Control), the curing stage (C3d and C5d), and the smoking stage (S5d and S10d). The results showed that LTS increased the hardness, transparency, and b* value of bacon fat. The increased contents of neutral lipids and free fatty acids, increased activities of neutral lipase and lipoxygenase, and increased peroxide and thiobarbituric acid reactive substance value indicated significant lipolysis and lipid oxidation of bacon fat during LTS. After the treatment, a decreased melting point and increased β'- and β-type fat crystal formation were observed in the fat portion. Moreover, the treatment led to disruption of the adipocyte membrane structure. Therefore, the destruction of adipocytes after lipolysis and lipid oxidation during low-temperature smoking treatment might contribute to the development of quality properties of bacon fat portions. Precise control of temperature and time enhances the stability of the fat portion of bacon, thereby improving quality characteristics such as texture and appearance.
Collapse
Affiliation(s)
- Chuxin Liu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Enqi He
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Peitao Fu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Leli He
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Lei Zhou
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Aihua Lou
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yan Liu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Haohua Fu
- Tangrenshen Group Co., Ltd., Zhuzhou 412000, China
| | - Qingwu Shen
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jie Luo
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Wei Quan
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
28
|
Zhang F, Ning Z, Wang C. Body roundness index and cognitive function in older adults: a nationwide perspective. Front Aging Neurosci 2024; 16:1466464. [PMID: 39634656 PMCID: PMC11616450 DOI: 10.3389/fnagi.2024.1466464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Background Obesity negatively impacts cognitive function. However, the correlation between the body roundness index (BRI) and cognitive performance remains inadequately explored. Methods This study used data from the National Health and Nutrition Examination Survey (NHANES) collected from 2011 to 2014 to examine the correlation between BRI and cognitive function in individuals aged 65 and older. Models of multiple linear regression were used to investigate the relationship between BRI and cognitive performance. Additionally, smoothed curve fittings explored potential non-linear associations. Interaction tests and subgroup analyses were also performed. Results One thousand eight hundred seventy participants were taken into account, revealing an important negative relationship between BRI levels and cognitive performance. In the fully adjusted model, elevated BRI was substantially correlated with lower Digit Symbol Substitution Test (DSST) scores (β = -0.34, 95% CI = -0.64 to -0.05, p = 0.023), indicating that the higher BRI values are linked to worse cognitive performance. Sex differences were observed, with males showing a stronger negative association (p for interaction = 0.040). Conclusion Elevated BRI is related to worse cognitive function in the elderly population.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhongxing Ning
- Department of Intensive Care Unit, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, China
| | - Can Wang
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
29
|
Frasca D, Romero M, Blomberg BB. Similarities in B Cell Defects between Aging and Obesity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1407-1413. [PMID: 39495900 DOI: 10.4049/jimmunol.2300670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 09/16/2024] [Indexed: 11/06/2024]
Abstract
The aging population is increasing worldwide, and there is also an increase in the aging population living with overweight and obesity, due to changes in lifestyle and in dietary patterns that elderly individuals experience later in life. Both aging and obesity are conditions of accelerated metabolic dysfunction and dysregulated immune responses. In this review, we summarize published findings showing that obesity induces changes in humoral immunity similar to those induced by aging and that the age-associated B cell defects are mainly due to metabolic changes. We discuss the role of the obese adipose tissue in inducing dysfunctional humoral responses and autoimmune Ab secretion.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
30
|
Souto RMCV, Corassa RB, Souto Júnior JV, Morais Neto OL. Prevalence of disability and associated functional limitations among older adults in Brazil. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003225. [PMID: 39541406 PMCID: PMC11563409 DOI: 10.1371/journal.pgph.0003225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Disabilities are a serious public health, social and human rights issue. Few studies address the relationship between disabilities and functioning among older adults. The study aimed to estimate the prevalence of disability and its' association with comorbidities and functional limitations in Brazilian elderly individuals. METHODS Data from the National Health Survey-PNS 2019 was used. Prevalence rates with its corresponding 95% confidence intervals (CI) were estimated for variables of interest. Chi-squared tests and multiple logistic regression were conducted to investigate associations and estimate crude and adjusted odds ratios (OR) using Stata 17.0 software. The critical value (p<0.05) was considered. RESULTS The overall prevalence of disability was 58.3% (95% CI 57.2-59.4). Moderate/severe disabilities accounted for 24.1 (95% CI 23.1-25.1) and was high among elderly people females (27.9%, 95% CI 26.5-29.3), unemployed (28.4%, 95% CI 27.3-29.6), with an income of up to one minimum wage (30.6%, 95% CI 29.1-32.2), lower education (28.7%, 95% CI 27.5-29.9) and not married (28.5%, 95% CI 27.1-29.9). Crude odds ratios of having functional limitations were 4.5 times higher among individuals with three or more comorbidities, and 32.5 times higher among those with two or more disabilities, compared to those without these conditions. CONCLUSION Having a disability is an important predictor of functional limitations, especially among women, and people with lower income and education. To address this problem, public health policies such as encouraging physical activity among the elderly in Brazil should be implemented.
Collapse
Affiliation(s)
| | - Rafael Belo Corassa
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | | | | |
Collapse
|
31
|
Zhang J, Guan X, Zhong X. Immunosenescence in digestive system cancers: Mechanisms, research advances, and therapeutic strategies. Semin Cancer Biol 2024; 106-107:234-250. [PMID: 39510149 DOI: 10.1016/j.semcancer.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
Increasing lifespans and external environmental factors have contributed to the increase of age-related diseases, particularly cancer. A decrease in immune surveillance and clearance of cancer cells is the result of immunosenescence, which involves the remodeling of immune organs, the changes and functional decline of immune cell subsets, in association with systemic low-grade chronic inflammation. Stem cells aging in bone marrow and thymic involution are the most important causes of immunosenescence. Senescent cancer cells promote the differentiation, recruitment, and functional upregulation of immune-suppressive cell subsets e.g. regulatory T cells (Tregs), myeloid-derived suppressor cell (MDSC), tumor-associated macrophages (TAMS) through senescence-associated secretory phenotype (SASP) further exacerbating the immunosuppressive microenvironment. For digestive system cancers, age-related damage to the intestinal mucosal barrier, the aging of gut-associated lymphoid tissue (GALT), exposure to xenobiotic stimuli throughout life, and dysbiosis make the local immune microenvironment more vulnerable. This article systematically reviews the research progress of immunosenescence and immune microenvironment in digestive system cancers, as well as the exploration of related therapy strategies, hoping to point out new directions for research in the digestive system cancers.
Collapse
Affiliation(s)
- Junyan Zhang
- Department of Surgical Oncology and General Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaojiao Guan
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Xinwen Zhong
- Department of Thoracic Surgery, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
32
|
Xie Y, Tang Y, Yang J, Atta M, Wang N, Qin H. Sesamol Alleviated Lipotoxicity-Induced Dysfunction in MIN6 Cells via Facilitating Cellular Senescence Caused by Endoplasmic Reticulum Stress. J Biochem Mol Toxicol 2024; 38:e70038. [PMID: 39470143 DOI: 10.1002/jbt.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/21/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Obesity is found to be a significant risk factor for type 2 diabetes mellitus (T2DM), attributed to lipotoxicity-induced β-cell dysfunction. However, the specific mechanism involved in the process remains incompletely unclarified. The current study demonstrated lipotoxicity resulted in the activation of ER stress, which increased the protein level of TXNIP, thereby inducing senescence-assiciated dysfunction in MIN6 cells under high fat environment. And we also found sesamol, a natural functional component extracted from sesame, was able to alleviate senescence-associated β-cell dysfunction induced by lipotoxicity by inhibiting ER stress and TXNIP. Our findings provided novel insights into senescence-related T2DM and propose innovative therapeutic approaches for utilizing sesamol in the treatment of T2DM in the obese elderly population.
Collapse
Affiliation(s)
- Yan Xie
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yongyan Tang
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jinxin Yang
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, China
| | - Mahnoor Atta
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, China
| | - Nan Wang
- Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hong Qin
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
33
|
Toba H, Takai S. Exploring the roles of SPARC as a proinflammatory factor and its potential as a novel therapeutic target against cardiovascular disease. Am J Physiol Heart Circ Physiol 2024; 327:H1174-H1186. [PMID: 39269452 DOI: 10.1152/ajpheart.00565.2024] [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: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Cardiovascular disease (CVD) is a leading cause of death worldwide, and the number of patients with CVD continues to increase despite extensive research and developments in this field. Chronic inflammation is a pivotal pathological component of CVD, and unveiling new proinflammatory factors will help devise novel preventive and therapeutic strategies. The extracellular matrix (ECM) not only provides structural support between cells but also contributes to cellular functions. Secreted protein acidic and rich in cysteine (SPARC) is a collagen-binding matricellular protein that is particularly induced during development and tissue remodeling. A proinflammatory role for SPARC has been demonstrated in various animal models, such as in the lipopolysaccharide-induced footpad model and dextran sodium sulfate-induced colitis model. Recent clinical studies reported a positive correlation between elevated plasma SPARC levels and hypertension, obesity, and the inflammatory marker high-sensitive C-reactive protein. In addition, SPARC gene deletion attenuates the cardiac injury induced by aging, myocardial infarction, and pressure load, suggesting that SPARC has deleterious effects on CVD. This review summarizes the regulatory and proinflammatory mechanisms of SPARC on CVD, chronic kidney disease (CKD), and cerebrovascular disease and discusses the rationale behind measuring SPARC as a biomarker of CVD and the effects of inhibition of SPARC in the prevention and treatment of CVD.
Collapse
Affiliation(s)
- Hiroe Toba
- Division of Pathological Sciences, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
- Department of Pharmacology, Educational Foundation of Osaka Medical and Pharmacological University, Takatsuki, Japan
| | - Shinji Takai
- Department of Pharmacology, Educational Foundation of Osaka Medical and Pharmacological University, Takatsuki, Japan
| |
Collapse
|
34
|
Wang T, Zhou D, Hong Z. Adipose tissue in older individuals: a contributing factor to sarcopenia. Metabolism 2024; 160:155998. [PMID: 39128607 DOI: 10.1016/j.metabol.2024.155998] [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: 05/05/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Sarcopenia is a geriatric syndrome characterized by a functional decline in muscle. The prevalence of sarcopenia increases with natural aging, becoming a serious health problem among elderly individuals. Therefore, understanding the pathology of sarcopenia is critical for inhibiting age-related alterations and promoting health and longevity in elderly individuals. The development of sarcopenia may be influenced by interactions between visceral and subcutaneous adipose tissue and skeletal muscle, particularly under conditions of chronic low-grade inflammation and metabolic dysfunction. This hypothesis is supported by the following observations: (i) accumulation of senescent cells in both adipose tissue and skeletal muscle with age; (ii) gut dysbiosis, characterized by an imbalance in gut microbial communities as the main trigger for inflammation, sarcopenia, and aged adipose tissue; and (iii) microbial dysbiosis, which could impact the onset or progression of a senescent state. Moreover, adipose tissue acts as an endocrine organ, releasing molecules that participate in intricate communication networks between organs. Our discussion focuses on novel adipokines and their role in regulating adipose tissue and muscle, particularly those influenced by aging and obesity, emphasizing their contributions to disease development. On the basis of these findings, we propose that age-related adipose tissue and sarcopenia are disorders characterized by chronic inflammation and metabolic dysregulation. Finally, we explore new potential therapeutic strategies involving specialized proresolving mediator (SPM) G protein-coupled receptor (GPCR) agonists, non-SPM GPCR agonists, transient receptor potential (TRP) channels, antidiabetic drugs in conjunction with probiotics and prebiotics, and compounds designed to target senescent cells and mitigate their pro-inflammatory activity.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| | - Dong Zhou
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
35
|
Liu Z, Xie W, Li H, Liu X, Lu Y, Lu B, Deng Z, Li Y. Novel perspectives on leptin in osteoarthritis: Focus on aging. Genes Dis 2024; 11:101159. [PMID: 39229323 PMCID: PMC11369483 DOI: 10.1016/j.gendis.2023.101159] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/10/2023] [Accepted: 09/16/2023] [Indexed: 09/05/2024] Open
Abstract
Osteoarthritis (OA) is a common chronic joint disease characterized by articular cartilage degeneration, subchondral sclerosis, synovitis, and osteophyte formation. OA is associated with disability and impaired quality of life, particularly among the elderly. Leptin, a 16-kD non-glycosylated protein encoded by the obese gene, is produced on a systemic and local basis in adipose tissue and the infrapatellar fat pad located in the knee. The metabolic mechanisms employed by leptin in OA development have been widely studied, with attention being paid to aging as a corroborative risk factor for OA. Hence, in this review, we have attempted to establish a potential link between leptin and OA, by focusing on aging-associated mechanisms and proposing leptin as a potential diagnostic and therapeutic target in aging-related mechanisms of OA that may provide fruitful guidance and emphasis for future research.
Collapse
Affiliation(s)
- Zimo Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xu Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhenhan Deng
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
36
|
dos Santos TW, Pereira QC, Fortunato IM, Oliveira FDS, Alvarez MC, Ribeiro ML. Body Composition and Senescence: Impact of Polyphenols on Aging-Associated Events. Nutrients 2024; 16:3621. [PMID: 39519454 PMCID: PMC11547493 DOI: 10.3390/nu16213621] [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: 09/09/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Aging is a dynamic and progressive process characterized by the gradual accumulation of cellular damage. The continuous functional decline in the intrinsic capacity of living organisms to precisely regulate homeostasis leads to an increased susceptibility and vulnerability to diseases. Among the factors contributing to these changes, body composition-comprised of fat mass and lean mass deposits-plays a crucial role in the trajectory of a disability. Particularly, visceral and intermuscular fat deposits increase with aging and are associated with adverse health outcomes, having been linked to the pathogenesis of sarcopenia. Adipose tissue is involved in the secretion of bioactive factors that can ultimately mediate inter-organ pathology, including skeletal muscle pathology, through the induction of a pro-inflammatory profile such as a SASP, cellular senescence, and immunosenescence, among other events. Extensive research has shown that natural compounds have the ability to modulate the mechanisms associated with cellular senescence, in addition to exhibiting anti-inflammatory, antioxidant, and immunomodulatory potential, making them interesting strategies for promoting healthy aging. In this review, we will discuss how factors such as cellular senescence and the presence of a pro-inflammatory phenotype can negatively impact body composition and lead to the development of age-related diseases, as well as how the use of polyphenols can be a functional measure for restoring balance, maintaining tissue quality and composition, and promoting health.
Collapse
Affiliation(s)
- Tanila Wood dos Santos
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Quélita Cristina Pereira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Isabela Monique Fortunato
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Fabrício de Sousa Oliveira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Marisa Claudia Alvarez
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Marcelo Lima Ribeiro
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| |
Collapse
|
37
|
Tavenier J, Nehlin JO, Houlind MB, Rasmussen LJ, Tchkonia T, Kirkland JL, Andersen O, Rasmussen LJH. Fisetin as a senotherapeutic agent: Evidence and perspectives for age-related diseases. Mech Ageing Dev 2024; 222:111995. [PMID: 39384074 DOI: 10.1016/j.mad.2024.111995] [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: 08/07/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
Fisetin, a flavonoid naturally occurring in plants, fruits, and vegetables, has recently gained attention for its potential role as a senotherapeutic agent for the treatment of age-related chronic diseases. Senotherapeutics target senescent cells, which accumulate with age and disease, in both circulating immune cell populations and solid organs and tissues. Senescent cells contribute to development of many chronic diseases, primarily by eliciting systemic chronic inflammation through their senescence-associated secretory phenotype. Here, we explore whether fisetin as a senotherapeutic can eliminate senescent cells, and thereby alleviate chronic diseases, by examining current evidence from in vitro studies and animal models that investigate fisetin's impact on age-related diseases, as well as from phase I/II trials in various patient populations. We discuss the application of fisetin in humans, including challenges and future directions. Our review of available data suggests that targeting senescent cells with fisetin offers a promising strategy for managing multiple chronic diseases, potentially transforming future healthcare for older and multimorbid patients. However, further studies are needed to establish the safety, pharmacokinetics, and efficacy of fisetin as a senotherapeutic, identify relevant and reliable outcome measures in human trials, optimize dosing, and better understand the possible limitations of fisetin as a senotherapeutic agent.
Collapse
Affiliation(s)
- Juliette Tavenier
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; The Hospital Pharmacy, Marielundsvej 25, Herlev 2730, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| | - Tamara Tchkonia
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N 2200, Denmark; The Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; Department of Psychology & Neuroscience, Duke University, 2020 West Main Street Suite 201, Durham, NC 27708, USA.
| |
Collapse
|
38
|
Lluch A, Latorre J, Oliveras-Cañellas N, Fernández-Sánchez A, Moreno-Navarrete JM, Castells-Nobau A, Comas F, Buxò M, Rodríguez-Hermosa JI, Ballester M, Espadas I, Martín-Montalvo A, Zhang B, Zhou Y, Burkhardt R, Höring M, Liebisch G, Castellanos-Rubio A, Santin I, Kar A, Laakso M, Pajukanta P, Olkkonen VM, Fernández-Real JM, Ortega FJ. A novel long non-coding RNA connects obesity to impaired adipocyte function. Mol Metab 2024; 90:102040. [PMID: 39362599 PMCID: PMC11544081 DOI: 10.1016/j.molmet.2024.102040] [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: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) can perform tasks of key relevance in fat cells, contributing, when defective, to the burden of obesity and its sequelae. Here, scrutiny of adipose tissue transcriptomes before and after bariatric surgery (GSE53378) granted identification of 496 lncRNAs linked to the obese phenotype. Only expression of linc-GALNTL6-4 displayed an average recovery over 2-fold and FDR-adjusted p-value <0.0001 after weight loss. The aim of the present study was to investigate the impact on adipocyte function and potential clinical value of impaired adipose linc-GALNTL6-4 in obese subjects. METHODS We employed transcriptomic analysis of public dataset GSE199063, and cross validations in two large transversal cohorts to report evidence of a previously unknown association of adipose linc-GALNTL6-4 with obesity. We then performed functional analyses in human adipocyte cultures, genome-wide transcriptomics, and untargeted lipidomics in cell models of loss and gain of function to explore the molecular implications of its associations with obesity and weight loss. RESULTS The expression of linc-GALNTL6-4 in human adipose tissue is adipocyte-specific and co-segregates with obesity, being normalized upon weight loss. This co-segregation is demonstrated in two longitudinal weight loss studies and two cross-sectional samples. While compromised expression of linc-GALNTL6-4 in obese subjects is primarily due to the inflammatory component in the context of obesity, adipogenesis requires the transcriptional upregulation of linc-GALNTL6-4, the expression of which reaches an apex in terminally differentiated adipocytes. Functionally, we demonstrated that the knockdown of linc-GALNTL6-4 impairs adipogenesis, induces alterations in the lipidome, and leads to the downregulation of genes related to cell cycle, while propelling in adipocytes inflammation, impaired fatty acid metabolism, and altered gene expression patterns, including that of apolipoprotein C1 (APOC1). Conversely, the genetic gain of linc-GALNTL6-4 ameliorated differentiation and adipocyte phenotype, putatively by constraining APOC1, also contributing to the metabolism of triglycerides in adipose. CONCLUSIONS Current data unveil the unforeseen connection of adipocyte-specific linc-GALNTL6-4 as a modulator of lipid homeostasis challenged by excessive body weight and meta-inflammation.
Collapse
Affiliation(s)
- Aina Lluch
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Jèssica Latorre
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.
| | - Núria Oliveras-Cañellas
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | | | - José M Moreno-Navarrete
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Anna Castells-Nobau
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain
| | - Ferran Comas
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain
| | - Maria Buxò
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain
| | - José I Rodríguez-Hermosa
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; School of Medicine, University of Girona (UdG), Girona, Spain
| | - María Ballester
- Animal Breeding and Genetics Programme, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, Caldes de Montbui, Spain
| | - Isabel Espadas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas (CSIC), University Pablo de Olavide, Seville, Spain
| | - Alejandro Martín-Montalvo
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas (CSIC), University Pablo de Olavide, Seville, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Birong Zhang
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - You Zhou
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ainara Castellanos-Rubio
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Izortze Santin
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Bizkaia, Spain; Instituto de Investigación Sanitaria Biocruces Bizkaia, Bizkaia, Spain
| | - Asha Kar
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles (CA), USA; Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles (CA), USA
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Päivi Pajukanta
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles (CA), USA; Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles (CA), USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles (CA), USA
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, University of Helsinki, Helsinki, Finland
| | - José M Fernández-Real
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain; School of Medicine, University of Girona (UdG), Girona, Spain.
| | - Francisco J Ortega
- Institut d'Investigació Biomèdica de Girona (IDIBGI) - Girona, Spain; CIBER de la Fisiología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.
| |
Collapse
|
39
|
Nishida C, Iemitsu M, Kurihara T, Kishigami K, Miyachi M, Sanada K. Differences in sarcopenia indices in elderly Japanese women and their relationships with obesity classified according to waist circumference, BMI, and body fat percentage. J Physiol Anthropol 2024; 43:22. [PMID: 39354553 PMCID: PMC11446072 DOI: 10.1186/s40101-024-00370-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/02/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Sarcopenic obesity (SO) is defined as a decrease in lean body mass and an increase in body fat mass (BFM) due to aging. Detecting SO in elderly women is important from the perspective of extending healthy life expectancy. While various indices of SO are currently used, there is no global consensus regarding diagnostic criteria for SO. This study aimed to examine the relationship between obesity indices (waist circumference (WC), body mass index (BMI), and body fat percentage (BFP)) and sarcopenia indices (total body muscle mass (TBM), appendicular lean mass (ALM), skeletal mass index (SMI)), and physical function (gait speed (GS), handgrip strength (HGS)). METHODS Subjects were 170 community-dwelling healthy elderly women aged 65-79 years (mean: 72.7 ± 5.78 years) who underwent measurements for WC, BMI, and BFP. A WC of ≥ 90cm was defined as the obese group, BMI was determined as weight (kg) divided by height squared (m2) and a cutoff of ≥ 25 kg/m2 was used to define the obesity group. BFM was measured using the bioelectrical impedance analysis (BIA) method and BFP was calculated from body weight and a cutoff of ≥ 30% was used to define the obesity group. TBM and ALM (kg) were measured using the BIA method, ALM (kg) was corrected for height (m2) to obtain SMI (kg/m2). Physical function was assessed by GS and HGS, which were measured by the 5-m walk test and a digital grip strength meter, respectively. RESULTS When obesity was assessed using BMI, WC and BFP, obese individuals had higher TBM, ALM and SMI, and lower GS among the sarcopenia indicators. HGS did not differ significantly between the non-obese and obese groups. CONCLUSION Our findings suggest HGS is thought to reflect muscle strength without being affected by obesity indices, suggesting that it may be useful in detecting possible sarcopenia in obese individuals.
Collapse
Affiliation(s)
- Chihiro Nishida
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Motoyuki Iemitsu
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Toshiyuki Kurihara
- Faculty of Science, Yamaguchi University, 3003 Yoshida, Yamaguchi, 753-8512, Japan
| | - Keiko Kishigami
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Motohiko Miyachi
- Faculty of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama, 359-1192, Japan
| | - Kiyoshi Sanada
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
40
|
Su R, Ai Y, Wang J, Wu L, Sun H, Ding M, Xie R, Liang Q. Engineered Microfibers for Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5823-5840. [PMID: 39145987 DOI: 10.1021/acsabm.4c00615] [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: 08/16/2024]
Abstract
Hydrogel microfibers are hydrogel materials engineered into fiber structures. Techniques such as wet spinning, microfluidic spinning, and 3D bioprinting are often used to prepare microfibers due to their ability to precisely control the size, morphology, and structure of the microfibers. Microfibers with different structural morphologies have different functions; they provide a flow-through culture environment for cells to improve viability, and can also be used to induce the differentiation of cells such as skeletal muscle and cardiac muscle cells to eventually form functional organs in vitro through special morphologies. This Review introduces recent advances in microfluidics, 3D bioprinting, and wet spinning in the preparation of microfibers, focusing on the materials and fabrication methods. The applications of microfibers in tissue engineering are highlighted by summarizing their contributions in engineering biomimetic blood vessels, vascularized tissues, bone, heart, pancreas, kidney, liver, and fat. Furthermore, applications of engineered fibers in tissue repair and drug screening are also discussed.
Collapse
Affiliation(s)
- Riguga Su
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Jingyu Wang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Lei Wu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Hua Sun
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Mingyu Ding
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Ruoxiao Xie
- Department of Materials, Design and Manufacturing Engineering, School of Engineering, University of Liverpool, Liverpool L69 3BX, U.K
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
41
|
Cortes TM, Vasquez L, Serra MC, Robbins R, Stepanenko A, Brown K, Barrus H, Campos A, Espinoza SE, Musi N. Effect of Semaglutide on Physical Function, Body Composition, and Biomarkers of Aging in Older Adults With Overweight and Insulin Resistance: Protocol for an Open-Labeled Randomized Controlled Trial. JMIR Res Protoc 2024; 13:e62667. [PMID: 39269759 PMCID: PMC11437224 DOI: 10.2196/62667] [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: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Older adults with type 2 diabetes mellitus (T2DM) or prediabetes are at increased risk of adverse changes in body composition, physical function, and aging-related biomarkers compared to those with normal glucose tolerance. Semaglutide is a glucagon-like peptide 1 receptor agonist that has been approved for T2DM and chronic weight management. Although semaglutide is effective for weight loss and T2DM management, its effects on lean body mass, physical function, and biomarkers of aging are understudied in older adults. OBJECTIVE This study aims to compare the effects of lifestyle counseling with and that without semaglutide on body composition, physical function, and biomarkers of aging in older adults. METHODS This is an open-label randomized controlled trial. A total of 20 adults (aged 65 years and older) with elevated BMI (27-40 kg/m2) and prediabetes or well-controlled T2DM (hemoglobin A1c 5.7%-7.5%) are recruited, stratified by sex, and randomized 1:1 to one of 2 groups (semaglutide plus lifestyle counseling vs lifestyle counseling alone) and followed up for 5 months. Those in the semaglutide group are titrated to 1 mg weekly, as tolerated, for 12 weeks. Lifestyle counseling is given by registered dietitians and based on the Diabetes Prevention Program Lifestyle Change Program. Our primary outcomes include changes in lean mass, physical function, and biomarkers of aging. Body composition is measured by dual-energy x-ray absorptiometry and includes total fat mass and lean mass. Physical function is measured by 6-minute walk distance, grip strength, and short physical performance battery. Biomarkers of aging are measured in blood, skeletal muscle, and abdominal adipose tissue to include C-reactive protein, interleukin-6, tumor necrosis factors α, and β galactosidase staining. RESULTS The study was funded in December 2021 with a projected data collection period from spring 2023 through summer 2024. CONCLUSIONS Despite the elevated risk of adverse changes in body composition, physical function, and biomarkers of aging among older adults with glucose intolerance and elevated adiposity, the benefits and risks of commonly prescribed antihyperglycemic or weight loss medications such as semaglutide are understudied. This study aims to fill this knowledge gap to inform clinicians about the potential for additional clinically meaningful, nonglycemic effects of semaglutide. TRIAL REGISTRATION ClinicalTrials.gov NCT05786521; https://clinicaltrials.gov/study/NCT05786521. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/62667.
Collapse
Affiliation(s)
- Tiffany M Cortes
- Division of Endocrinology, Department of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
- Sam & Ann Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
- San Antonio Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Libia Vasquez
- Texas Diabetes Institute, University Health System, San Antonio, TX, United States
| | - Monica C Serra
- Sam & Ann Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
- San Antonio Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, United States
- Division of Geriatrics, Gerontology & Palliative Medicine, Department of Medicine, University of Texas Health Science San Antonio, San Antonio, TX, United States
| | - Ronna Robbins
- Department of Nutrition and Food Science, Texas Woman's University, Denton, TX, United States
| | - Allison Stepanenko
- Sam & Ann Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
| | - Kevin Brown
- Sam & Ann Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
| | - Hannah Barrus
- Sam & Ann Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
| | - Annalisa Campos
- Sam & Ann Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
| | - Sara E Espinoza
- Division of Geriatrics, Gerontology & Palliative Medicine, Department of Medicine, University of Texas Health Science San Antonio, San Antonio, TX, United States
- Center for Translational Geroscience, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Diabetes and Aging Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Nicolas Musi
- Center for Translational Geroscience, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Diabetes and Aging Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
42
|
Suda M, Paul KH, Tripathi U, Minamino T, Tchkonia T, Kirkland JL. Targeting Cell Senescence and Senolytics: Novel Interventions for Age-Related Endocrine Dysfunction. Endocr Rev 2024; 45:655-675. [PMID: 38500373 PMCID: PMC11405506 DOI: 10.1210/endrev/bnae010] [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: 08/23/2023] [Revised: 01/11/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Multiple changes occur in hormonal regulation with aging and across various endocrine organs. These changes are associated with multiple age-related disorders and diseases. A better understanding of responsible underling biological mechanisms could help in the management of multiple endocrine disorders over and above hormone replacement therapy (HRT). Cellular senescence is involved in multiple biological aging processes and pathologies common in elderly individuals. Cellular senescence, which occurs in many older individuals but also across the lifespan in association with tissue damage, acute and chronic diseases, certain drugs, and genetic syndromes, may contribute to such endocrine disorders as osteoporosis, metabolic syndrome, and type 2 diabetes mellitus. Drugs that selectively induce senescent cell removal, "senolytics,", and drugs that attenuate the tissue-destructive secretory state of certain senescent cells, "senomorphics," appear to delay the onset of or alleviate multiple diseases, including but not limited to endocrine disorders such as diabetes, complications of obesity, age-related osteoporosis, and cancers as well as atherosclerosis, chronic kidney disease, neurodegenerative disorders, and many others. More than 30 clinical trials of senolytic and senomorphic agents have already been completed, are underway, or are planned for a variety of indications. Targeting senescent cells is a novel strategy that is distinct from conventional therapies such as HRT, and thus might address unmet medical needs and can potentially amplify effects of established endocrine drug regimens, perhaps allowing for dose decreases and reducing side effects.
Collapse
Affiliation(s)
- Masayoshi Suda
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Karl H Paul
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Utkarsh Tripathi
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| | - Tamara Tchkonia
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
43
|
Zhang Y, Jiang Y, Yang X, Huang Y, Pan A, Liao Y. Adipose tissue senescence: Biological changes, hallmarks and therapeutic approaches. Mech Ageing Dev 2024; 222:111988. [PMID: 39265709 DOI: 10.1016/j.mad.2024.111988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
Adipose tissue (AT), the largest energy storage reservoir and endocrine organ, plays a crucial role in regulating systemic energy metabolism. As one of the most vulnerable tissues during aging, the plasticity of AT is impaired. With age, AT undergoes redistribution, characterized by expansion of visceral adipose tissue (VAT) and reduction of peripheral subcutaneous adipose tissue (SAT). Additionally, age-related changes in AT include reduced adipogenesis of white adipocytes, decreased proliferation and differentiation capacity of mesenchymal stromal/stem cells (MSCs), diminished thermogenic capacity in brown/beige adipocytes, and dysregulation of immune cells. Specific and sensitive hallmarks enable the monitoring and evaluation of the biological changes associated with aging. In this study, we have innovatively proposed seven characteristic hallmarks of AT senescence, including telomere attrition, epigenetic alterations, genomic instability, mitochondrial dysfunction, disabled macroautophagy, cellular senescence, and chronic inflammation, which are intricately interconnected and mutually regulated. Finally, we discussed anti-aging strategies targeting AT, offering insights into mitigating or delaying metabolic disturbances caused by AT senescence.
Collapse
Affiliation(s)
- Yajuan Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Yaoyao Jiang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Xiaoyue Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Yumei Huang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China.
| |
Collapse
|
44
|
Theodorou SJ, Theodorou DJ, Kigka V, Gkiatas I, Fotopoulos A. DXA-based appendicular composition measures in healthy aging Caucasian Greek women: a cross-sectional study. Rheumatol Int 2024; 44:1715-1723. [PMID: 38860993 DOI: 10.1007/s00296-024-05622-4] [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: 02/11/2024] [Accepted: 05/26/2024] [Indexed: 06/12/2024]
Abstract
As the global population of older persons increases, age-related medical conditions will have a greater impact on public health. DXA-derived bone and soft tissue metrics are associated with adverse clinical events in aging persons. This study aims to investigate the regional body composition of the appendices by whole-body DXA scans, and the age-related relationships between measures of bone and soft tissue in healthy Caucasian females of a Greek origin residing in the Mediterranean area. Body composition of the legs and the arms was analyzed, and lean mass (LM) and fat mass (FM) metrics were calculated in 330 women aged 20-85 years, using DXA. Peak bone mineral density (BMD) of the legs and arms was achieved between ages 20-30 and 41-50 years, respectively. The overall BMD reduction with age was for the legs 43% and the arms 32.2% (p < 0.001). Peak %LM of the legs and the arms was achieved between ages 20-30. The overall reduction of %LM with age was for the legs 22.5% (p < 0.001) and arms 6.6% (p < 0.05). Peak %FM of the legs and arms was attained between ages 31-40 and 61-70, respectively. The overall %FM reduction with age was for the legs and arms 7.5% and 1.9% (p > 0.05). In appendicular sites, Greek women reach peak values of bone mass in the legs first, in early adulthood. Bone loss predominates in the legs as women age. Also, with advancing age Greek women show preferential significant decreases of %LM and %FM in the legs as opposed to the arms. Although variation in appendicular bone and soft tissue metrics is present, the implications of variable biological crosstalks among the tissue components as women age may ultimately lay the foundation for future clinical trials aimed at healthy aging.
Collapse
Affiliation(s)
| | - Daphne J Theodorou
- Department of Radiology, General Hospital of Ioannina and National Healthcare System, Ioannina, Greece
| | - Vassiliki Kigka
- Department of Orthopaedic Surgery, University of Ioannina, Ioannina, Greece
| | - Ioannis Gkiatas
- Department of Orthopaedic Surgery, University of Ioannina, Ioannina, Greece
| | - Andreas Fotopoulos
- Department of Nuclear Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
45
|
Turkoglu B, Mansuroglu B. Investigating the Effects of Chelidonic Acid on Oxidative Stress-Induced Premature Cellular Senescence in Human Skin Fibroblast Cells. Life (Basel) 2024; 14:1070. [PMID: 39337855 PMCID: PMC11433492 DOI: 10.3390/life14091070] [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: 07/30/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
This study investigated the effects of chelidonic acid (CA) on hydrogen peroxide (H2O2) induced cellular senescence in human skin fibroblast cells (BJ). Cellular senescence is a critical mechanism that is linked to age-related diseases and chronic conditions. CA, a γ-pyrone compound known for its broad pharmacological activity, was assessed for its potential to mitigate oxidative stress and alter senescence markers. A stress-induced premature senescence (SIPS) model was designed in BJ fibroblast cells using the oxidative stress agent H2O2. After this treatment, cells were treated with CA, and the potential effect of CA on senescence was evaluated using senescence-related β-galactosidase, 4',6-diamino-2-phenylindole (DAPI), acridine-orange staining (AO), comet assay, molecular docking assays, gene expression, and protein analysis. These results demonstrate that CA effectively reduces senescence markers, including senescence-associated β-galactosidase activity, DNA damage, lysosomal activity, and oxidative stress indicators such as malondialdehyde. Molecular docking revealed CA's potential interactions with critical proteins involved in senescence signalling pathways, suggesting mechanisms by which CA may exert its effects. Gene expression and protein analyses corroborated the observed anti-senescent effects, with CA modulating p16, p21, and pRB1 expressions and reducing oxidative stress markers. In conclusion, CA appeared to have senolytic and senomorphic potential in vitro, which could mitigate and reverse SIPS markers in BJ fibroblasts.
Collapse
Affiliation(s)
| | - Banu Mansuroglu
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Yildiz Technical University, Istanbul 34220, Turkey;
| |
Collapse
|
46
|
Wang S, Wang R, Hu Y, Zhang Y, Yuan Q, Luo Y, Yuan C. Long noncoding RNA AI504432 upregulates FASN expression by sponging miR-1a-3p to promote lipogenesis in senescent adipocytes. Cell Signal 2024; 120:111232. [PMID: 38763183 DOI: 10.1016/j.cellsig.2024.111232] [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: 12/29/2023] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Aging affects lipid metabolism and can cause obesity as it is closely related to the disorder of many lipogenic regulatory factors. LncRNAs have been recognized as pivotal regulators across diverse biological processes, but their effects on lipogenesis in aging remain to be further studied. In this work, using RNA sequencing (RNA-Seq), we found that the expression of lncRNA AI504432 was significantly upregulated in the eWAT (epididymal white adipose tissue) of aging mice, and the knockdown of AI504432 notably reduced the expression of several adipogenic genes (e.g., Cebp/α, Srebp-1c, Fasn, Acaca, and Scd1) in senescent adipocytes. The bioinformatics investigation revealed that AI504432 possessed a binding site for miR-1a-3p, and the discovery was verified by the luciferase reporter assay. The expression of Fasn was increased upon the inhibition of miR-1a-3p but restored upon the simultaneous silencing of AI504432. Taken together, our results suggested that AI504432 controlled lipogenesis through the miR-1a-3p/Fasn signaling pathway. The findings may inspire new therapeutic approaches to target imbalanced lipid homeostasis due to aging.
Collapse
Affiliation(s)
- Shuwen Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Rui Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yaqi Hu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yifan Zhang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Qi Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China; College of Medicine and Health Science, China Three Gorges University, Yichang 443002, China
| | - Yiyang Luo
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China; College of Medicine and Health Science, China Three Gorges University, Yichang 443002, China
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
47
|
Xia M, Li W, Lin H, Zeng H, Ma S, Wu Q, Ma H, Li X, Pan B, Gao J, Hu Y, Liu Y, Wang S, Gao X. DNA methylation age acceleration contributes to the development and prediction of non-alcoholic fatty liver disease. GeroScience 2024; 46:3525-3542. [PMID: 37605101 PMCID: PMC11226581 DOI: 10.1007/s11357-023-00903-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/06/2023] [Indexed: 08/23/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is prevalent in the aging society. Despite body weight reduction, the prevalence of NAFLD has been increasing with aging for unknown reasons. Here, we investigate the association of DNA methylation age acceleration, a hallmark of aging, with risk of NAFLD. Genome-wide DNA methylation profiles were measured in 95 participants who developed type 2 diabetes during 4-year follow-up, and 356 randomly sampled participants from Shanghai Changfeng Study. DNA methylation age was calculated using the Horvath's method, and liver fat content (LFC) was measured using a quantitative ultrasound method. Subjects with highest tertile of DNA methylation age acceleration (≥ 9.5 years) had significantly higher LFC (7.2% vs 3.1%, P = 0.008) but lower body fat percentage (29.7% vs 33.0%, P = 0.032) than those with lowest tertile of DNA methylation age acceleration (< 4.0 years). After adjustment for age, sex, alcohol drinking, cigarette smoking, BMI, waist circumference, and different type blood cell counts, the risk of NAFLD was still significantly increased in the highest tertile group (OR, 4.55; 95% CI, 1.06-19.61). Even in subjects with similar LFC at baseline, DNA methylation age acceleration was associated with higher increase in LFC (4.0 ± 10.7% vs 0.9 ± 9.5%, P = 0.004) after a median of 4-year follow-up. Further analysis found that 6 CpGs of Horvath age predictors were associated with longitudinal changes in LFC after multivariate adjustment and located on genes that might lead to fat redistribution from peripheral adipose to liver. Combination of the key CpG methylation related to liver fat content with conventional risk factors improves the performance for NAFLD prediction.
Collapse
Affiliation(s)
- Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Wenran Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Rd, Shanghai, 200031, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Hailuan Zeng
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Shuai Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Qi Wu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Hui Ma
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoming Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian Gao
- Department of Nutrition, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Yu Hu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Rd, Shanghai, 200031, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China.
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
48
|
Qi N, Franczyk MP, Yamaguchi S, Kojima D, Hayashi K, Satoh A, Ogiso N, Kanda T, Sasaki Y, Finck BN, DeBosch BJ, Yoshino J. Adipocyte-specific inactivation of NAMPT, a key NAD + biosynthetic enzyme, causes a metabolically unhealthy lean phenotype in female mice during aging. Am J Physiol Endocrinol Metab 2024; 327:E81-E88. [PMID: 38809511 PMCID: PMC11390120 DOI: 10.1152/ajpendo.00313.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a universal coenzyme regulating cellular energy metabolism in many cell types. Recent studies have demonstrated the close relationships between defective NAD+ metabolism and aging and age-associated metabolic diseases. The major purpose of the present study was to test the hypothesis that NAD+ biosynthesis, mediated by a rate-limiting NAD+ biosynthetic enzyme, nicotinamide phosphoribosyltransferase (NAMPT), is essential for maintaining normal adipose tissue function and whole body metabolic health during the aging process. To this end, we provided in-depth and comprehensive metabolic assessments for female adipocyte-specific Nampt knockout (ANKO) mice during aging. We first evaluated body fat mass in young (≤4-mo-old), middle aged (10-14-mo-old), and old (≥18-mo-old) mice. Intriguingly, adipocyte-specific Nampt deletion protected against age-induced obesity without changing energy balance. However, data obtained from the hyperinsulinemic-euglycemic clamp procedure (HECP) demonstrated that, despite the lean phenotype, old ANKO mice had severe insulin resistance in skeletal muscle, heart, and white adipose tissue (WAT). Old ANKO mice also exhibited hyperinsulinemia and hypoadiponectinemia. Mechanistically, loss of Nampt caused marked decreases in WAT gene expression of lipogenic targets of peroxisome proliferator-activated receptor gamma (PPAR-γ) in an age-dependent manner. In addition, administration of a PPAR-γ agonist rosiglitazone restored fat mass and improved metabolic abnormalities in old ANKO mice. In conclusion, these findings highlight the importance of the NAMPT-NAD+-PPAR-γ axis in maintaining functional integrity and quantity of adipose tissue, and whole body metabolic function in female mice during aging.NEW & NOTEWORTHY Defective NAD+ metabolism is associated with aging and age-associated metabolic diseases. In the present study, we provided in-depth metabolic assessments in female mice with adipocyte-specific inactivation of a key NAD+ biosynthetic enzyme NAMPT and revealed an unexpected role of adipose tissue NAMPT-NAD+-PPAR-γ axis in maintaining functional integrity and quantity of adipose tissue and whole body metabolic health during the aging process.
Collapse
Affiliation(s)
- Nathan Qi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Michael P Franczyk
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Shintaro Yamaguchi
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, United States
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Daiki Kojima
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kaori Hayashi
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akiko Satoh
- Department of Integrative Physiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
- Department of Integrative Physiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Noboru Ogiso
- Laboratory of Experimental Animals, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takeshi Kanda
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan
- The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Japan
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Brian N Finck
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Brian J DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jun Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, United States
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan
- The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Japan
| |
Collapse
|
49
|
Lee MC, Chung YC, Thenaka PC, Wang YW, Lin YL, Kan NW. Effects of different HIIT protocols on exercise performance, metabolic adaptation, and fat loss in middle-aged and older adults with overweight. Int J Med Sci 2024; 21:1689-1700. [PMID: 39006847 PMCID: PMC11241097 DOI: 10.7150/ijms.96073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/04/2024] [Indexed: 07/16/2024] Open
Abstract
Introduction: There is evidence that aging and obesity are associated with increased oxidative stress and chronic inflammation. High-intensity interval training (HIIT) may be superior to moderate-intensity continuous training (MICT) in anti-inflammatory and anti-obesity benefits. Therefore, the objective of this study is to determine which HIIT prescriptions will be more effective in reducing fat accumulation, inflammation, and improving metabolic adaptation and exercise performance in middle-aged and older overweight adults. Methods: Thirty-six middle-aged with overweight adults were divided into one of three groups: 1. L-HIIT group: the long-interval HIIT group (4 × 4 min Exercise/4 min Rest), 2. M-HIIT group: the medium-interval HIIT group (8 × 2 min Exercise/2 min Rest), 3. Control group: no exercise training intervention. All groups underwent the training stage for eight weeks (three sessions per week), followed by a detraining stage of four weeks in order to investigate the effects induced by different HIIT interventions on inflammation, metabolic adaptation, anti-fatigue and exercise performance, and fat loss Results: There was a significant physiological response in the change rate of heart rate (HR) after an acute L-HIIT session compared with an acute M-HIIT session (ΔHR: ↑49.66±16.09% vs ↑33.22±14.37%, p=0.02); furthermore, systolic blood pressure (SBP) and diastolic blood pressure (DBP) decreased significantly following a single L-HIIT session. After an eight-week training stage, the L-HIIT and M-HIIT groups exhibited a significant increase in aerobic capacity (ΔVO2peak), with values of +27.93±16.79% (p<0.001) and +18.39±8.12% (p<0.001), respectively, in comparison to the control group. Furthermore, in the L-HIIT group, the anaerobic power of relative mean power (RMP) exhibited a significant increase (p=0.019). However, following a four-week detraining stage, the adiponectin concentration remained 1.78 times higher in the L-HIIT group than in the control group (p=0.033). The results of blood sugar, blood lipids, body composition, and inflammatory markers did not indicate any improved it did not indicate any improvements from the two different HIIT protocols. Conclusions: The results indicate that an eight-week L-HIIT or M-HIIT intervention (three sessions per week, 32 minutes per session) may be an effective approach for improving aerobic capacity. It can be posited that L-HIIT may be a more advantageous mode than M-HIIT for enhancing anaerobic power, adipokine levels, and improving blood pressure in an aged and overweight population due to the induced physiological responses.
Collapse
Affiliation(s)
- Mon-Chien Lee
- Center for General Education, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan, Taiwan
| | - Yu-Chun Chung
- Center for General Education, Taipei Medical University, Taipei, Taiwan
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Priskila Cherisca Thenaka
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei, Taiwan
| | - Ya-Wen Wang
- Center for General Education, Taipei Medical University, Taipei, Taiwan
| | - Yea-Lih Lin
- Center for General Education, Taipei Medical University, Taipei, Taiwan
| | - Nai-Wen Kan
- Center for General Education, Taipei Medical University, Taipei, Taiwan
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
50
|
Xia B, Zeng P, Xue Y, Li Q, Xie J, Xu J, Wu W, Yang X. Identification of potential shared gene signatures between gastric cancer and type 2 diabetes: a data-driven analysis. Front Med (Lausanne) 2024; 11:1382004. [PMID: 38903804 PMCID: PMC11187270 DOI: 10.3389/fmed.2024.1382004] [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: 02/04/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
Background Gastric cancer (GC) and type 2 diabetes (T2D) contribute to each other, but the interaction mechanisms remain undiscovered. The goal of this research was to explore shared genes as well as crosstalk mechanisms between GC and T2D. Methods The Gene Expression Omnibus (GEO) database served as the source of the GC and T2D datasets. The differentially expressed genes (DEGs) and weighted gene co-expression network analysis (WGCNA) were utilized to identify representative genes. In addition, overlapping genes between the representative genes of the two diseases were used for functional enrichment analysis and protein-protein interaction (PPI) network. Next, hub genes were filtered through two machine learning algorithms. Finally, external validation was undertaken with data from the Cancer Genome Atlas (TCGA) database. Results A total of 292 and 541 DEGs were obtained from the GC (GSE29272) and T2D (GSE164416) datasets, respectively. In addition, 2,704 and 336 module genes were identified in GC and T2D. Following their intersection, 104 crosstalk genes were identified. Enrichment analysis indicated that "ECM-receptor interaction," "AGE-RAGE signaling pathway in diabetic complications," "aging," and "cellular response to copper ion" were mutual pathways. Through the PPI network, 10 genes were identified as candidate hub genes. Machine learning further selected BGN, VCAN, FN1, FBLN1, COL4A5, COL1A1, and COL6A3 as hub genes. Conclusion "ECM-receptor interaction," "AGE-RAGE signaling pathway in diabetic complications," "aging," and "cellular response to copper ion" were revealed as possible crosstalk mechanisms. BGN, VCAN, FN1, FBLN1, COL4A5, COL1A1, and COL6A3 were identified as shared genes and potential therapeutic targets for people suffering from GC and T2D.
Collapse
Affiliation(s)
- Bingqing Xia
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping Zeng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuling Xue
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Jiamin Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Wenzhen Wu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Xiaobo Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|