1
|
Kieronska-Rudek A, Ascencao K, Chlopicki S, Szabo C. Increased hydrogen sulfide turnover serves a cytoprotective role during the development of replicative senescence. Biochem Pharmacol 2024; 230:116595. [PMID: 39454733 DOI: 10.1016/j.bcp.2024.116595] [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: 05/06/2024] [Revised: 09/15/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
The mammalian gasotransmitter hydrogen sulfide (H2S) is produced by enzymes such as cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), 3-mercaptopyruvate sulfurtransferase (3-MST). Prior studies suggest that H2S may have cytoprotective and anti-aging effects. This project explores the regulation and role of endogenous H2S in a murine model of replicative senescence. H2S and polysulfide levels in RAW 264.7 murine macrophages (control cells: passage 5-10; senescent cells: passage 30-40) were measured using fluorescent probes. The expression of H2S-related enzymes and the activity of senescence marker beta-galactosidase (SA-β-Gal) were also analyzed. CBS, CSE, and 3-MST were inhibited using selective pharmacological inhibitors. Senescence led to a moderate upregulation of CBS and in a significant increase in CSE and 3-MST. H2S degradation enzymes were also elevated in senescence. Inhibition of H2S-producing enzymes reduced H2S levels but increased polysulfides. Inhibition of H2S production during senescence suppressed cell proliferation, and elevated SA-β-Gal and p21 levels. Comparing young and old mice spleens revealed downregulation of CBS and ETHE1 and upregulation of rhodanese and SUOX in older mice. The results demonstrate that increased reactive sulfur turnover occurs in senescent macrophages and that reactive sulfur species support cell proliferation and regulate cellular senescence.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Cracow, Poland
| | - Kelly Ascencao
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Cracow, Poland; Jagiellonian University Medical College, Chair of Pharmacology, Faculty of Medicine, Cracow, Poland
| | - Csaba Szabo
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
2
|
Liu Y, Fang M, Tu X, Mo X, Zhang L, Yang B, Wang F, Kim YB, Huang C, Chen L, Fan S. Dietary Polyphenols as Anti-Aging Agents: Targeting the Hallmarks of Aging. Nutrients 2024; 16:3305. [PMID: 39408272 PMCID: PMC11478989 DOI: 10.3390/nu16193305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/20/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Aging is a natural biological process influenced by multiple factors and is a significant contributor to various chronic diseases. Slowing down the aging process and extending health span have been pursuits of the scientific field. Methods: Examination of the effects of dietary polyphenols on hallmarks of aging such as genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. Results: Polyphenols, abundant in nature, exhibit numerous biological activities, including antioxidant effects, free radical scavenging, neuroprotection, and anti-aging properties. These compounds are generally safe and effective in potentially slowing aging and preventing age-related disorders. Conclusions: The review encourages the development of novel therapeutic strategies using dietary polyphenols to create holistic anti-aging therapies and nutritional supplements.
Collapse
Affiliation(s)
- Ying Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.L.); (C.H.)
| | - Minglv Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.L.); (C.H.)
| | - Xiaohui Tu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.L.); (C.H.)
| | - Xueying Mo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.L.); (C.H.)
| | - Lu Zhang
- Nutrilite Health Institute, Amway (Shanghai) Innovation and Science Co., Ltd., Shanghai 201203, China
| | - Binrui Yang
- Nutrilite Health Institute, Amway (Shanghai) Innovation and Science Co., Ltd., Shanghai 201203, China
| | - Feijie Wang
- Nutrilite Health Institute, Amway (Shanghai) Innovation and Science Co., Ltd., Shanghai 201203, China
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.L.); (C.H.)
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation and Science Co., Ltd., Shanghai 201203, China
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.L.); (C.H.)
| |
Collapse
|
3
|
Gao H, Nepovimova E, Adam V, Heger Z, Valko M, Wu Q, Kuca K. Age-associated changes in innate and adaptive immunity: role of the gut microbiota. Front Immunol 2024; 15:1421062. [PMID: 39351234 PMCID: PMC11439693 DOI: 10.3389/fimmu.2024.1421062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Aging is generally regarded as an irreversible process, and its intricate relationship with the immune system has garnered significant attention due to its profound implications for the health and well-being of the aging population. As people age, a multitude of alterations occur within the immune system, affecting both innate and adaptive immunity. In the realm of innate immunity, aging brings about changes in the number and function of various immune cells, including neutrophils, monocytes, and macrophages. Additionally, certain immune pathways, like the cGAS-STING, become activated. These alterations can potentially result in telomere damage, the disruption of cytokine signaling, and impaired recognition of pathogens. The adaptive immune system, too, undergoes a myriad of changes as age advances. These include shifts in the number, frequency, subtype, and function of T cells and B cells. Furthermore, the human gut microbiota undergoes dynamic changes as a part of the aging process. Notably, the interplay between immune changes and gut microbiota highlights the gut's role in modulating immune responses and maintaining immune homeostasis. The gut microbiota of centenarians exhibits characteristics akin to those found in young individuals, setting it apart from the microbiota observed in typical elderly individuals. This review delves into the current understanding of how aging impacts the immune system and suggests potential strategies for reversing aging through interventions in immune factors.
Collapse
Affiliation(s)
- Haoyu Gao
- College of Life Science, Yangtze University, Jingzhou, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| |
Collapse
|
4
|
Greenberg EF, Voorbach MJ, Smith A, Reuter DR, Zhuang Y, Wang JQ, Wooten DW, Asque E, Hu M, Hoft C, Duggan R, Townsend M, Orsi K, Dalecki K, Amberg W, Duggan L, Knight H, Spina JS, He Y, Marsh K, Zhao V, Ybarra S, Mollon J, Fang Y, Vasanthakumar A, Westmoreland S, Droescher M, Finnema SJ, Florian H. Navitoclax safety, tolerability, and effect on biomarkers of senescence and neurodegeneration in aged nonhuman primates. Heliyon 2024; 10:e36483. [PMID: 39253182 PMCID: PMC11382177 DOI: 10.1016/j.heliyon.2024.e36483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Alzheimer's disease (AD) is the most common global dementia and is universally fatal. Most late-stage AD disease-modifying therapies are intravenous and target amyloid beta (Aβ), with only modest effects on disease progression: there remains a high unmet need for convenient, safe, and effective therapeutics. Senescent cells (SC) and the senescence-associated secretory phenotype (SASP) drive AD pathology and increase with AD severity. Preclinical senolytic studies have shown improvements in neuroinflammation, tau, Aβ, and CNS damage; most were conducted in transgenic rodent models with uncertain human translational relevance. In this study, aged cynomolgus monkeys had significant elevation of biomarkers of senescence, SASP, and neurological damage. Intermittent treatment with the senolytic navitoclax induced modest reversible thrombocytopenia; no serious drug-related toxicity was noted. Navitoclax reduced several senescence and SASP biomarkers, with CSF concentrations sufficient for senolysis. Finally, navitoclax reduced TSPO-PET frontal cortex binding and showed trends of improvement in CSF biomarkers of neuroinflammation, neuronal damage, and synaptic dysfunction. Overall, navitoclax administration was safe and well tolerated in aged monkeys, inducing trends of biomarker changes relevant to human neurodegenerative disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Min Hu
- AbbVie Inc., North Chicago, IL, United States
| | - Carolin Hoft
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Ryan Duggan
- AbbVie Inc., North Chicago, IL, United States
| | - Matthew Townsend
- AbbVie, Cambridge Research Center, 200 Sidney Street, Cambridge, MA, 02139, United States
| | - Karin Orsi
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | | | - Willi Amberg
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Lori Duggan
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Heather Knight
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Joseph S Spina
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Yupeng He
- AbbVie Inc., North Chicago, IL, United States
| | | | - Vivian Zhao
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | - Suzanne Ybarra
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | - Jennifer Mollon
- AbbVie Deutschland GmbH & Co. KG, Statistical Sciences and Analytics, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Yuni Fang
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | | | - Susan Westmoreland
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Mathias Droescher
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | | | | |
Collapse
|
5
|
Sun S, Jiang M, Ma S, Ren J, Liu GH. Exploring the heterogeneous targets of metabolic aging at single-cell resolution. Trends Endocrinol Metab 2024:S1043-2760(24)00190-5. [PMID: 39181730 DOI: 10.1016/j.tem.2024.07.009] [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: 04/23/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 08/27/2024]
Abstract
Our limited understanding of metabolic aging poses major challenges to comprehending the diverse cellular alterations that contribute to age-related decline, and to devising targeted interventions. This review provides insights into the heterogeneous nature of cellular metabolism during aging and its response to interventions, with a specific focus on cellular heterogeneity and its implications. By synthesizing recent findings using single-cell approaches, we explored the vulnerabilities of distinct cell types and key metabolic pathways. Delving into the cell type-specific alterations underlying the efficacy of systemic interventions, we also discuss the complexity of integrating single-cell data and advocate for leveraging computational tools and artificial intelligence to harness the full potential of these data, develop effective strategies against metabolic aging, and promote healthy aging.
Collapse
Affiliation(s)
- Shuhui Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China.
| | - Mengmeng Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Shuai Ma
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium, Beijing 100101, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium, Beijing 100101, China; Key Laboratory of RNA Innovation, Science and Engineering, China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guang-Hui Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
6
|
Liang Y, Kaushal D, Wilson RB. Cellular Senescence and Extracellular Vesicles in the Pathogenesis and Treatment of Obesity-A Narrative Review. Int J Mol Sci 2024; 25:7943. [PMID: 39063184 PMCID: PMC11276987 DOI: 10.3390/ijms25147943] [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: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
This narrative review explores the pathophysiology of obesity, cellular senescence, and exosome release. When exposed to excessive nutrients, adipocytes develop mitochondrial dysfunction and generate reactive oxygen species with DNA damage. This triggers adipocyte hypertrophy and hypoxia, inhibition of adiponectin secretion and adipogenesis, increased endoplasmic reticulum stress and maladaptive unfolded protein response, metaflammation, and polarization of macrophages. Such feed-forward cycles are not resolved by antioxidant systems, heat shock response pathways, or DNA repair mechanisms, resulting in transmissible cellular senescence via autocrine, paracrine, and endocrine signaling. Senescence can thus affect preadipocytes, mature adipocytes, tissue macrophages and lymphocytes, hepatocytes, vascular endothelium, pancreatic β cells, myocytes, hypothalamic nuclei, and renal podocytes. The senescence-associated secretory phenotype is closely related to visceral adipose tissue expansion and metaflammation; inhibition of SIRT-1, adiponectin, and autophagy; and increased release of exosomes, exosomal micro-RNAs, pro-inflammatory adipokines, and saturated free fatty acids. The resulting hypernefemia, insulin resistance, and diminished fatty acid β-oxidation lead to lipotoxicity and progressive obesity, metabolic syndrome, and physical and cognitive functional decline. Weight cycling is related to continuing immunosenescence and exposure to palmitate. Cellular senescence, exosome release, and the transmissible senescence-associated secretory phenotype contribute to obesity and metabolic syndrome. Targeted therapies have interrelated and synergistic effects on cellular senescence, obesity, and premature aging.
Collapse
Affiliation(s)
- Yicong Liang
- Bankstown Hospital, University of New South Wales, Sydney, NSW 2560, Australia;
| | - Devesh Kaushal
- Campbelltown Hospital, Western Sydney University, Sydney, NSW 2560, Australia;
| | - Robert Beaumont Wilson
- School of Clinical Medicine, University of New South Wales, High St., Kensington, Sydney, NSW 2052, Australia
| |
Collapse
|
7
|
Yusri K, Kumar S, Fong S, Gruber J, Sorrentino V. Towards Healthy Longevity: Comprehensive Insights from Molecular Targets and Biomarkers to Biological Clocks. Int J Mol Sci 2024; 25:6793. [PMID: 38928497 PMCID: PMC11203944 DOI: 10.3390/ijms25126793] [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: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Aging is a complex and time-dependent decline in physiological function that affects most organisms, leading to increased risk of age-related diseases. Investigating the molecular underpinnings of aging is crucial to identify geroprotectors, precisely quantify biological age, and propose healthy longevity approaches. This review explores pathways that are currently being investigated as intervention targets and aging biomarkers spanning molecular, cellular, and systemic dimensions. Interventions that target these hallmarks may ameliorate the aging process, with some progressing to clinical trials. Biomarkers of these hallmarks are used to estimate biological aging and risk of aging-associated disease. Utilizing aging biomarkers, biological aging clocks can be constructed that predict a state of abnormal aging, age-related diseases, and increased mortality. Biological age estimation can therefore provide the basis for a fine-grained risk stratification by predicting all-cause mortality well ahead of the onset of specific diseases, thus offering a window for intervention. Yet, despite technological advancements, challenges persist due to individual variability and the dynamic nature of these biomarkers. Addressing this requires longitudinal studies for robust biomarker identification. Overall, utilizing the hallmarks of aging to discover new drug targets and develop new biomarkers opens new frontiers in medicine. Prospects involve multi-omics integration, machine learning, and personalized approaches for targeted interventions, promising a healthier aging population.
Collapse
Affiliation(s)
- Khalishah Yusri
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sanjay Kumar
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sheng Fong
- Department of Geriatric Medicine, Singapore General Hospital, Singapore 169608, Singapore
- Clinical and Translational Sciences PhD Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jan Gruber
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Science Division, Yale-NUS College, Singapore 138527, Singapore
| | - Vincenzo Sorrentino
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Neuroscience Cellular & Molecular Mechanisms, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
8
|
Nguyen TT, Corvera S. Adipose tissue as a linchpin of organismal ageing. Nat Metab 2024; 6:793-807. [PMID: 38783156 PMCID: PMC11238912 DOI: 10.1038/s42255-024-01046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/10/2024] [Indexed: 05/25/2024]
Abstract
Ageing is a conserved biological process, modulated by intrinsic and extrinsic factors, that leads to changes in life expectancy. In humans, ageing is characterized by greatly increased prevalence of cardiometabolic disease, type 2 diabetes and disorders associated with impaired immune surveillance. Adipose tissue displays species-conserved, temporal changes with ageing, including redistribution from peripheral to central depots, loss of thermogenic capacity and expansion within the bone marrow. Adipose tissue is localized to discrete depots, and also diffusely distributed within multiple organs and tissues in direct proximity to specialized cells. Thus, through their potent endocrine properties, adipocytes are capable of modulating tissue and organ function throughout the body. In addition to adipocytes, multipotent progenitor/stem cells in adipose tissue play a crucial role in maintenance and repair of tissues throughout the lifetime. Adipose tissue may therefore be a central driver for organismal ageing and age-associated diseases. Here we review the features of adipose tissue during ageing, and discuss potential mechanisms by which these changes affect whole-body metabolism, immunity and longevity. We also explore the potential of adipose tissue-targeted therapies to ameliorate age-associated disease burdens.
Collapse
Affiliation(s)
- Tammy T Nguyen
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center, Worcester, MA, USA
- Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - Silvia Corvera
- Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA.
- Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, USA.
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
9
|
Balducci L, Falandry C, Silvio Monfardini. Senotherapy, cancer, and aging. J Geriatr Oncol 2024; 15:101671. [PMID: 37977898 DOI: 10.1016/j.jgo.2023.101671] [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: 09/28/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION We aimed to highlight the effects of senotherapy on the prevention and treatment of cancer in older individuals. The aim of senotherapy is to eliminate senescent cells. These cells express the senescence-associated secretory phenotype (SASP). With production of inflammatory cytokines, growth factors, and different type of proteases, the SASP is responsible for aging-associated disability and diseases. All mammalian cells experience senescence. The main agents of aging include fibroblasts and adipose cells. Senescent tumor cells may undergo genomic reprogramming and re-enter cell cycle with a stem cell phenotype. MATERIALS AND METHODS We conducted a Medline search for the following key words: senotherapy, senolysis, senomorphic agents. We provide a narrative review of the finding. RESULTS Different agents may eliminate senescent cells from cell cultures and murine models. These include metformin, rapamycin, desatinib, quercitin, fisetin, ruloxitinib, and BCL2 inhibitors. A randomized controlled study of metformin in 3,000 patients aged 65-79 without glucose intolerance aiming to establish whether senotherapy may prevent or reverse disability and aging associated diseases, including cancer, is ongoing. Senotherapy prolongs the life span and decreases the incidence of cancer in experimental animal models, as well as delays and reverses disability. Senescent tumor cells are found prior to treatment and after chemotherapy and radiation. These elements may be responsible for tumor recurrence and treatment refractoriness. DISCUSSION Senotherapy may have substantial effects on cancer management including decreased incidence and aggressiveness of cancer, improved tolerance of antineoplastic treatment, and prevention of relapse after primary treatment. Senotherapy may ameliorate several complications of cancer chemotherapy.
Collapse
Affiliation(s)
| | - Claire Falandry
- Service de Gériatrie, Centre Hospitaliser Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Laboratoire CarMeN, Inserm U1060, INRA U1397, Université Claude Bernard Lyon, France.
| | - Silvio Monfardini
- Director Oncopaedia Project European School of Oncology. Director Emeritus Division of Medical Oncology Istituto Oncologico Veneto, Padova., Italy.
| |
Collapse
|
10
|
Hall SA, Lesniewski LA. Targeting vascular senescence in cardiovascular disease with aging. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:16. [PMID: 39119148 PMCID: PMC11309369 DOI: 10.20517/jca.2023.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Aging is a major risk factor for atherosclerosis and cardiovascular disease (CVD). Two major age-associated arterial phenotypes, endothelial dysfunction and large elastic arterial stiffness, are autonomous predictors of future CVD diagnosis and contribute to the progression of CVD in older adults. Senescent cells lose the capacity to proliferate but remain metabolically active and secrete inflammatory factors termed senescence-associated secretory phenotype (SASP), leading to an increase in inflammation and oxidative stress. Accumulation of senescent cells is linked with the progression of age-related diseases and has been known to play a role in cardiovascular disease. In this brief review, we describe the characteristics and mechanisms of senescent cell accumulation and how senescent cells promote endothelial dysfunction and arterial stiffness. We focus on a range of novel therapeutic strategies aimed at reducing the burden of endothelial dysfunction leading to atherosclerosis through targeting senescent cells. Studies have begun to investigate a specific class of drugs that are able to selectively eliminate senescent cells, termed senolytics, which have shown great promise in reversing the aging phenotype and ameliorating pathologies in age-related disorders, creating a new opportunity for aging research. Generating therapies targeting the elimination of senescent cells would improve health span and increase longevity, making senolytics a promising therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Shelby A Hall
- Department of Nutrition and Integrated Physiology, University of Utah, Salt Lake City, UT 84112, USA
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Lisa A Lesniewski
- Department of Nutrition and Integrated Physiology, University of Utah, Salt Lake City, UT 84112, USA
- Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA
- Geriatric Research Education and Clinical Centers, Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
11
|
Sharma R. Exploring the emerging bidirectional association between inflamm-aging and cellular senescence in organismal aging and disease. Cell Biochem Funct 2024; 42:e3970. [PMID: 38456500 DOI: 10.1002/cbf.3970] [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: 01/03/2024] [Revised: 02/15/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
There is strong evidence that most individuals in the elderly population are characterized by inflamm-aging which refers to a subtle increase in the systemic pro-inflammatory environment and impaired innate immune activation. Although a variety of distinct factors are associated with the progression of inflamm-aging, emerging research is demonstrating a dynamic relationship between the processes of cellular senescence and inflamm-aging. Cellular senescence is a recognized factor governing organismal aging, and through a characteristic secretome, accumulating senescent cells can induce and augment a pro-inflammatory tissue environment that provides a rationale for immune system-independent activation of inflamm-aging and associated diseases. There is also accumulating evidence that inflamm-aging or its components can directly accelerate the development of senescent cells and ultimately senescent cell burden in tissues in a likely vicious inflammatory loop. The present review is intended to describe the emerging senescence-based molecular etiology of inflamm-aging as well as the dynamic reciprocal interactions between inflamm-aging and cellular senescence. Therapeutic interventions concurrently targeting cellular senescence and inflamm-aging are discussed and limitations as well as research opportunities have been deliberated. An effort has been made to provide a rationale for integrating inflamm-aging with cellular senescence both as an underlying cause and therapeutic target for further studies.
Collapse
Affiliation(s)
- Rohit Sharma
- Nutrigerontology Laboratory, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India
| |
Collapse
|
12
|
Lee E, Carreras-Gallo N, Lopez L, Turner L, Lin A, Mendez TL, Went H, Tomusiak A, Verdin E, Corley M, Ndhlovu L, Smith R, Dwaraka VB. Exploring the effects of Dasatinib, Quercetin, and Fisetin on DNA methylation clocks: a longitudinal study on senolytic interventions. Aging (Albany NY) 2024; 16:3088-3106. [PMID: 38393697 PMCID: PMC10929829 DOI: 10.18632/aging.205581] [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: 08/03/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
Senolytics, small molecules targeting cellular senescence, have emerged as potential therapeutics to enhance health span. However, their impact on epigenetic age remains unstudied. This study aimed to assess the effects of Dasatinib and Quercetin (DQ) senolytic treatment on DNA methylation (DNAm), epigenetic age, and immune cell subsets. In a Phase I pilot study, 19 participants received DQ for 6 months, with DNAm measured at baseline, 3 months, and 6 months. Significant increases in epigenetic age acceleration were observed in first-generation epigenetic clocks and mitotic clocks at 3 and 6 months, along with a notable decrease in telomere length. However, no significant differences were observed in second and third-generation clocks. Building upon these findings, a subsequent investigation evaluated the combination of DQ with Fisetin (DQF), a well-known antioxidant and antiaging senolytic molecule. After one year, 19 participants (including 10 from the initial study) received DQF for 6 months, with DNAm assessed at baseline and 6 months. Remarkably, the addition of Fisetin to the treatment resulted in non-significant increases in epigenetic age acceleration, suggesting a potential mitigating effect of Fisetin on the impact of DQ on epigenetic aging. Furthermore, our analyses unveiled notable differences in immune cell proportions between the DQ and DQF treatment groups, providing a biological basis for the divergent patterns observed in the evolution of epigenetic clocks. These findings warrant further research to validate and comprehensively understand the implications of these combined interventions.
Collapse
Affiliation(s)
- Edwin Lee
- Institute For Hormonal Balance, Orlando, FL 32819, USA
| | | | | | | | - Aaron Lin
- TruDiagnostic, Lexington, KY 40503, USA
| | | | | | - Alan Tomusiak
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | | | | |
Collapse
|
13
|
Nieto M, Konigsberg M, Silva-Palacios A. Quercetin and dasatinib, two powerful senolytics in age-related cardiovascular disease. Biogerontology 2024; 25:71-82. [PMID: 37747577 DOI: 10.1007/s10522-023-10068-5] [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: 07/17/2023] [Accepted: 09/01/2023] [Indexed: 09/26/2023]
Abstract
Cellular senescence is characteristic of the development and progression of multiple age-associated diseases. Accumulation of senescent cells in the heart contributes to various age-related pathologies. Several compounds called senolytics have been designed to eliminate these cells within the tissues. In recent years, the use and study of senolytics increased, representing a promising field for finding accessible and safe therapies for cardiovascular disease (CVD) treatment. This mini-review discusses the changes in the aging heart and the participation of senescent cells in CVD, as well as the use of senolytics to prevent the progression of myocardial damage, mainly the effect of dasatinib and quercetin. In particular, the mechanisms and physiological effects of senolytics therapies in the aged heart are discussed.
Collapse
Affiliation(s)
- Mario Nieto
- Department of Cardiovascular Biomedicine, National Institute of Cardiology, Ignacio Chávez, Juan Badiano No. 1. Colonia Sección XVI, 14080, Mexico City, Mexico
- Department of Health Sciences, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Mina Konigsberg
- Department of Health Sciences, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, National Institute of Cardiology, Ignacio Chávez, Juan Badiano No. 1. Colonia Sección XVI, 14080, Mexico City, Mexico.
| |
Collapse
|
14
|
Rex N, Melk A, Schmitt R. Cellular senescence and kidney aging. Clin Sci (Lond) 2023; 137:1805-1821. [PMID: 38126209 PMCID: PMC10739085 DOI: 10.1042/cs20230140] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Life expectancy is increasing worldwide, and by 2050 the proportion of the world's population over 65 years of age is estimated to surpass 1.5 billion. Kidney aging is associated with molecular and physiological changes that cause a loss of renal function and of regenerative potential. As the aging population grows, it is crucial to understand the mechanisms underlying these changes, as they increase the susceptibility to developing acute kidney injury (AKI) and chronic kidney disease (CKD). Various cellular processes and molecular pathways take part in the complex process of kidney aging. In this review, we will focus on the phenomenon of cellular senescence as one of the involved mechanisms at the crossroad of kidney aging, age-related disease, and CKD. We will highlight experimental and clinical findings about the role of cellular senescence in kidney aging and CKD. In addition, we will review challenges in senescence research and emerging therapeutic aspects. We will highlight the great potential of senolytic strategies for the elimination of harmful senescent cells to promote healthy kidney aging and to avoid age-related disease and CKD. This review aims to give insight into recent discoveries and future developments, providing a comprehensive overview of current knowledge on cellular senescence and anti-senescent therapies in the kidney field.
Collapse
Affiliation(s)
- Nikolai Rex
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
| | - Anette Melk
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Germany
| | - Roland Schmitt
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|