51
|
Tomimatsu N, Di Cristofaro LFM, Kanji S, Samentar L, Jordan BR, Kittler R, Habib AA, Espindola-Netto JM, Tchkonia T, Kirkland JL, Burns TC, Sarkaria JN, Gilbert A, Floyd JR, Hromas R, Zhao W, Zhou D, Sung P, Mukherjee B, Burma S. Targeting cIAP2 in a novel senolytic strategy prevents glioblastoma recurrence after radiotherapy. EMBO Mol Med 2025; 17:645-678. [PMID: 39972068 PMCID: PMC11982261 DOI: 10.1038/s44321-025-00201-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: 07/09/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Glioblastomas (GBM) are routinely treated with high doses of ionizing radiation (IR), yet these tumors recur quickly, and the recurrent tumors are highly therapy resistant. Here, we report that IR-induced senescence of tumor cells counterintuitively spurs GBM recurrence, driven by the senescence-associated secretory phenotype (SASP). We find that irradiated GBM cell lines and patient derived xenograft (PDX) cultures senesce rapidly in a p21-dependent manner. Senescent glioma cells upregulate SASP genes and secrete a panoply of SASP factors, prominently interleukin IL-6, an activator of the JAK-STAT3 pathway. These SASP factors collectively activate the JAK-STAT3 and NF-κB pathways in non-senescent GBM cells, thereby promoting tumor cell proliferation and SASP spreading. Transcriptomic analyses of irradiated GBM cells and the TCGA database reveal that the cellular inhibitor of apoptosis protein 2 (cIAP2), encoded by the BIRC3 gene, is a potential survival factor for senescent glioma cells. Senescent GBM cells not only upregulate BIRC3 but also induce BIRC3 expression and promote radioresistance in non-senescent tumor cells. We find that second mitochondria-derived activator of caspases (SMAC) mimetics targeting cIAP2 act as novel senolytics that trigger apoptosis of senescent GBM cells with minimal toxicity towards normal brain cells. Finally, using both PDX and immunocompetent mouse models of GBM, we show that the SMAC mimetic birinapant, administered as an adjuvant after radiotherapy, can eliminate senescent GBM cells and prevent the emergence of recurrent tumors. Taken together, our results clearly indicate that significant improvement in GBM patient survival may become possible in the clinic by eliminating senescent cells arising after radiotherapy.
Collapse
Affiliation(s)
- Nozomi Tomimatsu
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | | | - Suman Kanji
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | - Lorena Samentar
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | - Benjamin Russell Jordan
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amyn A Habib
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | | | - Terry C Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Andrea Gilbert
- Department of Pathology, University of Texas Health, San Antonio, TX, USA
| | - John R Floyd
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA
| | - Robert Hromas
- Department of Medicine, University of Texas Health, San Antonio, TX, USA
| | - Weixing Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Patrick Sung
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA
| | - Bipasha Mukherjee
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA.
| | - Sandeep Burma
- Department of Neurosurgery, University of Texas Health, San Antonio, TX, USA.
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA.
| |
Collapse
|
52
|
Babakhani K, Kucinskas AL, Ye X, Giles ED, Sun Y. Aging immunity: unraveling the complex nexus of diet, gut microbiome, and immune function. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00061. [PMID: 40352822 PMCID: PMC12063687 DOI: 10.1097/in9.0000000000000061] [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] [Received: 12/13/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
Aging is associated with immune senescence and gut dysbiosis, both of which are heavily influenced by the diet. In this review, we summarize current knowledge regarding the impact of diets high in fiber, protein, or fat, as well as different dietary components (tryptophan, omega-3 fatty acids, and galacto-oligosaccharides) on the immune system and the gut microbiome in aging. Additionally, this review discusses how aging alters tryptophan metabolism, contributing to changes in immune function and the gut microbiome. Understanding the relationship between diet, the gut microbiome, and immune function in the context of aging is critical to formulate sound dietary recommendations for older individuals, and these personalized nutritional practices will ultimately improve the health and longevity of the elderly.
Collapse
Affiliation(s)
| | - Amanda L. Kucinskas
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Xiangcang Ye
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| |
Collapse
|
53
|
Pignolo RJ, Chandra A. Insights into age-related osteoporosis from senescence-based preclinical models and human accelerated aging paradigms. Mech Ageing Dev 2025; 224:112025. [PMID: 39805505 PMCID: PMC11938943 DOI: 10.1016/j.mad.2025.112025] [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: 10/30/2024] [Revised: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Preclinical models of age-related osteoporosis have been developed based on the accumulation and clearance of senescent cells. The former include animal models based on telomere dysfunction and focal radiation; the latter based on genetic and pharmacological targeting (i.e., removal) of senescent cells. The weight of evidence using these models suggests that cellular senescence plays a key role in the pathophysiology of aging-onset bone loss with the senescence-associated secretory phenotype (SASP) mediating local and systemic deleterious effects on the skeleton. Mitochondrial dysfunction has also been implicated in senescence and age-related comorbidities, including osteoporosis, and knock-in mutations in the mtDNA polymerase gamma (Polg) gene in mice may recapitulate similar respiratory chain complex defects in aged individual with osteoporosis. This and other contributions to senile osteoporosis may also be identified by the careful evaluation of non-genetic paradigms of human accelerated aging. Premature aging syndromes, especially those with a prominent bone loss phenotype, include clinical scenarios of skeletal unloading, premature ovarian failure and survival from childhood cancers. These non-hereditary progeroid syndromes implicate the involvement of lineage switching to an adipogenic fate, inhibition of Wnt signaling, increased osteoclastogenesis and activation frequency of osteoclasts, as well as the substantial burden of senescent cell accumulation.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of Medicine, Divisions of Geriatric Medicine and Gerontology, Endocrinology, and Hospital Internal Medicine, the Department of Physiology and Biomedical Engineering, and the Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| | - Abhishek Chandra
- Department of Medicine, Divisions of Geriatric Medicine and Gerontology, the Department of Physiology and Biomedical Engineering, and the Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
54
|
Tilton M, Liao J, Kim C, Shaygani H, Potes MA, Cordova DJ, Kirkland JL, Miller KM. Tracing Cellular Senescence in Bone: Time-Dependent Changes in Osteocyte Cytoskeleton Mechanics and Morphology. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408517. [PMID: 40026102 PMCID: PMC11985287 DOI: 10.1002/smll.202408517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/22/2025] [Indexed: 03/04/2025]
Abstract
Aging-related bone loss significantly impacts the growing elderly population globally, leading to debilitating conditions such as osteoporosis. Senescent osteocytes play a crucial role in the aging process of bone. This longitudinal study examines the impact of continuous local and paracrine exposure to senescence-associated secretory phenotype (SASP) factors on biophysical and biomolecular markers in osteocytes. Significant cytoskeletal stiffening in irradiated (IR) osteocytes are found, accompanied by expansion of F-actin areas and a decline in dendritic integrity. These changes, correlating with alterations in pro-inflammatory cytokine levels and osteocyte-specific gene expression, support the reliability of biophysical markers for identifying senescent osteocytes. Notably, local accumulation of SASP factors have a more pronounced impact on osteocyte biophysical properties than paracrine effects, suggesting that the interplay between local and paracrine exposure can substantially influence cellular aging. This study underscores the importance of osteocyte mechanical and morphological properties as biophysical markers of senescence, highlighting their time dependence and differential effects of local and paracrine SASP exposure. Collectively, the investigation into biophysical senescence markers offers unique and reliable functional hallmarks for the non-invasive identification of senescent osteocytes, providing insights that can inform therapeutic strategies to mitigate aging-related bone loss.
Collapse
Affiliation(s)
- Maryam Tilton
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Junhan Liao
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chanul Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hossein Shaygani
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Maria Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Domenic J. Cordova
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kyle M. Miller
- Department of Radiation Oncology Emory University School of Medicine Atlanta, GA 30307, USA
| |
Collapse
|
55
|
Poulios P, Skampouras S, Piperi C. Deciphering the role of cytokines in aging: Biomarker potential and effective targeting. Mech Ageing Dev 2025; 224:112036. [PMID: 39832637 DOI: 10.1016/j.mad.2025.112036] [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: 11/06/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Aging is often characterized by chronic inflammation, immune system dysregulation, and cellular senescence with chronically elevated levels of pro-inflammatory cytokines. These small glycoproteins are mainly secreted by immune cells, mediating intercellular communication and immune system modulation through inflammatory signaling. Their pro- and anti-inflammatory effects make them a noteworthy research topic as well as a promising ally in combating inflammation and the aging process. Cytokines exert a synergistic role in aging and disease and may prove useful biomarkers of tissue-specific dysregulation, disease diagnosis and monitoring, presenting potential therapeutic options as anti-inflammatory and senolytic medications. In this review, we address the cellular and molecular mechanisms implicating cytokines in the aging process and related diseases, highlighting their biomarker potential. We focus on the current therapeutic strategies, including specific pharmaceutical agents, supplements, a balanced diet, and healthy habits such as exercise, stress management, and caloric restriction.
Collapse
Affiliation(s)
- Panagiotis Poulios
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Stamoulis Skampouras
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Christina Piperi
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens 11527, Greece.
| |
Collapse
|
56
|
Fang Z, Raza U, Song J, Lu J, Yao S, Liu X, Zhang W, Li S. Systemic aging fuels heart failure: Molecular mechanisms and therapeutic avenues. ESC Heart Fail 2025; 12:1059-1080. [PMID: 39034866 PMCID: PMC11911610 DOI: 10.1002/ehf2.14947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024] Open
Abstract
Systemic aging influences various physiological processes and contributes to structural and functional decline in cardiac tissue. These alterations include an increased incidence of left ventricular hypertrophy, a decline in left ventricular diastolic function, left atrial dilation, atrial fibrillation, myocardial fibrosis and cardiac amyloidosis, elevating susceptibility to chronic heart failure (HF) in the elderly. Age-related cardiac dysfunction stems from prolonged exposure to genomic, epigenetic, oxidative, autophagic, inflammatory and regenerative stresses, along with the accumulation of senescent cells. Concurrently, age-related structural and functional changes in the vascular system, attributed to endothelial dysfunction, arterial stiffness, impaired angiogenesis, oxidative stress and inflammation, impose additional strain on the heart. Dysregulated mechanosignalling and impaired nitric oxide signalling play critical roles in the age-related vascular dysfunction associated with HF. Metabolic aging drives intricate shifts in glucose and lipid metabolism, leading to insulin resistance, mitochondrial dysfunction and lipid accumulation within cardiomyocytes. These alterations contribute to cardiac hypertrophy, fibrosis and impaired contractility, ultimately propelling HF. Systemic low-grade chronic inflammation, in conjunction with the senescence-associated secretory phenotype, aggravates cardiac dysfunction with age by promoting immune cell infiltration into the myocardium, fostering HF. This is further exacerbated by age-related comorbidities like coronary artery disease (CAD), atherosclerosis, hypertension, obesity, diabetes and chronic kidney disease (CKD). CAD and atherosclerosis induce myocardial ischaemia and adverse remodelling, while hypertension contributes to cardiac hypertrophy and fibrosis. Obesity-associated insulin resistance, inflammation and dyslipidaemia create a profibrotic cardiac environment, whereas diabetes-related metabolic disturbances further impair cardiac function. CKD-related fluid overload, electrolyte imbalances and uraemic toxins exacerbate HF through systemic inflammation and neurohormonal renin-angiotensin-aldosterone system (RAAS) activation. Recognizing aging as a modifiable process has opened avenues to target systemic aging in HF through both lifestyle interventions and therapeutics. Exercise, known for its antioxidant effects, can partly reverse pathological cardiac remodelling in the elderly by countering processes linked to age-related chronic HF, such as mitochondrial dysfunction, inflammation, senescence and declining cardiomyocyte regeneration. Dietary interventions such as plant-based and ketogenic diets, caloric restriction and macronutrient supplementation are instrumental in maintaining energy balance, reducing adiposity and addressing micronutrient and macronutrient imbalances associated with age-related HF. Therapeutic advancements targeting systemic aging in HF are underway. Key approaches include senomorphics and senolytics to limit senescence, antioxidants targeting mitochondrial stress, anti-inflammatory drugs like interleukin (IL)-1β inhibitors, metabolic rejuvenators such as nicotinamide riboside, resveratrol and sirtuin (SIRT) activators and autophagy enhancers like metformin and sodium-glucose cotransporter 2 (SGLT2) inhibitors, all of which offer potential for preserving cardiac function and alleviating the age-related HF burden.
Collapse
Affiliation(s)
- Zhuyubing Fang
- Cardiovascular Department of Internal MedicineKaramay Hospital of People's Hospital of Xinjiang Uygur Autonomous RegionKaramayXinjiang Uygur Autonomous RegionChina
| | - Umar Raza
- School of Basic Medical SciencesShenzhen UniversityShenzhenGuangdong ProvinceChina
| | - Jia Song
- Department of Medicine (Cardiovascular Research)Baylor College of MedicineHoustonTexasUSA
| | - Junyan Lu
- Department of CardiologyZengcheng Branch of Nanfang Hospital, Southern Medical UniversityGuangzhouGuangdong ProvinceChina
| | - Shun Yao
- Department of NeurosurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouGuangdong ProvinceChina
| | - Xiaohong Liu
- Cardiovascular Department of Internal MedicineKaramay Hospital of People's Hospital of Xinjiang Uygur Autonomous RegionKaramayXinjiang Uygur Autonomous RegionChina
| | - Wei Zhang
- Outpatient Clinic of SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouGuangdong ProvinceChina
| | - Shujuan Li
- Department of Pediatric CardiologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouGuangdong ProvinceChina
| |
Collapse
|
57
|
Alcon C, Kovatcheva M, Morales-Sánchez P, López-Polo V, Torres T, Puig S, Lu A, Samitier J, Enrich C, Serrano M, Montero J. HRK downregulation and augmented BCL-xL binding to BAK confer apoptotic protection to therapy-induced senescent melanoma cells. Cell Death Differ 2025; 32:646-656. [PMID: 39627361 PMCID: PMC11982230 DOI: 10.1038/s41418-024-01417-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 04/11/2025] Open
Abstract
Senescent cells are commonly detected in tumors after chemo and radiotherapy, leading to a characteristic cellular phenotype that resists apoptotic cell death. In this study, we used multiple melanoma cell lines, molecular markers, and therapies to investigate the key role of the BCL-2 family proteins in the survival of senescent cells. We first used BH3 profiling to assess changes in apoptotic priming upon senescence induction. Unexpectedly, not all cell types analyzed showed a decrease in apoptotic priming, BIM was downregulated, there was variability in BAX expression and BAK remained constant or increased. Therefore, there was not a clear pattern for pro-survival adaptation. Many studies have been devoted to find ways to eliminate senescent cells, leading to one of the most studied senolytic agents: navitoclax, a promiscuous BH3 mimetic that inhibits BCL-2, BCL-xL and BCL-W. While it is known that the BCL-2 family of proteins is commonly upregulated in senescent cells, the complexity of the apoptotic network has not been fully explored. Interestingly, we found distinct protein expression changes always leading to a BCL-xL mediated pro-survival adaptation, as assessed by BH3 profiling. When analyzing potential therapeutic strategies, we observed a stronger senolytic activity in these melanoma cell lines when specifically targeting BCL-xL using A-1331852, navitoclax or the PROTAC BCL-xL degrader DT2216. We found that the sensitizer protein HRK was systematically downregulated when senescence was induced, leading to an increased availability of BCL-xL. Furthermore, we identified that the main apoptotic inhibition was shaped by BCL-xL and BAK binding increase that prevented mitochondrial permeabilization and apoptosis. To our knowledge, this is the first time that the molecular basis for BCL-xL anti-apoptotic adaptation in senescence is described, paving the way for the development of new molecules that either prevent HRK downregulation or displace BCL-xL binding to BAK to be used as senolytics.
Collapse
Affiliation(s)
- Clara Alcon
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| | - Marta Kovatcheva
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- IFOM, The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Paula Morales-Sánchez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Vanessa López-Polo
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Teresa Torres
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Susana Puig
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
- Dermatology Department, Hospital Clinic and Fundació Clínic per la Recerca Biomèdica., Barcelona, Spain
| | - Albert Lu
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Department of Electronics and Biomedical Engineering, Faculty of Physics, University of Barcelona, Barcelona, Spain
| | - Carlos Enrich
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | - Joan Montero
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
58
|
Luna A, Chou K, Wragg KM, Worley MJ, Paruchuri N, Zhou X, Blin MG, Moore BB, Salmon M, Goldstein DR, Deng JC. Senolytic treatment attenuates immune cell infiltration without improving IAV outcomes in aged mice. Aging Cell 2025; 24:e14437. [PMID: 39754380 PMCID: PMC11984683 DOI: 10.1111/acel.14437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/06/2025] Open
Abstract
Aging is a major risk factor for poor outcomes following respiratory infections. In animal models, the most severe outcomes of respiratory infections in older hosts have been associated with an increased burden of senescent cells that accumulate over time with age and create a hyperinflammatory response. Although studies using coronavirus animal models have demonstrated that removal of senescent cells with senolytics, a class of drugs that selectively kills senescent cells, resulted in reduced lung damage and increased survival, little is known about the role that senescent cells play in the outcome of influenza A viral (IAV) infections in aged mice. Here, we tested if the aged mice survival or weight loss IAV infections could be improved using three different senolytic regimens. We found that neither dasatinib plus quercetin, fisetin, nor ABT-263 improved outcomes. Furthermore, both dasatanib plus quercetin and fisetin treatments further suppressed immune infiltration than aging alone. Additionally, our data show that the short-term senolytic agents do not reduce senescent markers in our aged mouse model. These findings suggest that acute senolytic treatments do not universally reverse aging related immune phenotype against all respiratory viral infections.
Collapse
Affiliation(s)
- Adrian Luna
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Kai‐Neng Chou
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Kathleen M. Wragg
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Matthew J. Worley
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Nikhil Paruchuri
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Xiaofeng Zhou
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Muriel G. Blin
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Bethany B. Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Morgan Salmon
- Department of Cardiac SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel R. Goldstein
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Jane C. Deng
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Veterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
| |
Collapse
|
59
|
Sangfuang N, McCoubrey LE, Awad A, Marzorati M, Ghyselinck J, Verstrepen L, Munck JD, Medts JD, Gaisford S, Basit AW. Effects of senotherapeutics on gut microbiome dysbiosis and intestinal inflammation in Crohn's disease: A pilot study. Transl Res 2025; 278:36-47. [PMID: 39986536 DOI: 10.1016/j.trsl.2025.02.004] [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: 05/30/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Inflammatory Bowel Disease (IBD) is characterized by chronic inflammation in the gastrointestinal tract, and is usually accompanied by dysbiosis in the gut microbiome, a factor that contributes to disease progression. Excessive production of reactive oxygen species (ROS) because of gut microbiome dysbiosis-one of the hallmark features of IBD-promotes chronic inflammation and facilitates the transformation of normal cells into senescent cells. Cellular senescence is associated with the development of various chronic and age-related diseases. We hypothesise that senolytic agents, specifically dasatinib (D) and quercetin (Q), could have a beneficial effect on both the gut microbiome and intestinal cells in IBD. The modulatory effects of a combination of D + Q was assessed in the M-SHIME model with faecal microbiota sourced from Crohn's disease patients. D + Q significantly modulated butyrate and lactate levels in the samples from specific patients. In addition, metabolomic analysis showed that D + Q positively impacted the abundance of anti-inflammatory bacteria while also significantly reducing the several species of pathogenic bacteria. Findings from a Caco-2 cell/THP1 co-culture model of IBD demonstrated that D + Q exerted strong immunomodulatory effects on the gut epithelium, evidenced by reduced NF-kB activity, and lower levels of the pro-inflammatory markers TNF-α, CXCL-10, and MCP-1. Furthermore, D + Q induced the secretion of anti-inflammatory cytokines, including IL-6 and IL-10. However, it should be noted that D + Q also led to the secretion of the pro-inflammatory cytokines IL-8. These findings suggest that D + Q could offer a novel therapeutic approach for advanced IBD management by modulating both the gut microbiome and inflammatory pathways. The results support the potential repurposing of senotherapeutic agents as a strategy for addressing the chronic inflammation central to IBD pathogenesis.
Collapse
Affiliation(s)
| | - Laura E McCoubrey
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK; Now at Drug Product Development, GSK R&D, Ware SG12 0GX, UK
| | - Atheer Awad
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK; Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, UK
| | | | | | | | | | | | - Simon Gaisford
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
60
|
Rattanaprukskul K, Xia XJ, Hysa M, Jiang M, Hung M, Suslavich S, Sahingur S. Dasatinib and Quercetin Limit Gingival Senescence, Inflammation, and Bone Loss. J Dent Res 2025; 104:419-427. [PMID: 39797437 PMCID: PMC11909784 DOI: 10.1177/00220345241299789] [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] [Indexed: 01/13/2025] Open
Abstract
Cellular senescence has emerged as one of the central hallmarks of aging and drivers of chronic comorbidities, including periodontal diseases. Senescence can also occur in younger tissues and instigate metabolic alterations and dysfunction, culminating in accelerated aging and pathological consequences. Senotherapeutics, such as the combination of dasatinib and quercetin (DQ), are being increasingly used to improve the clinical outcomes of chronic disorders and promote a healthy life span through the reduction of senescent cell burden and senescence-associated secretory phenotype (SASP). Recent evidence suggests that senescent cells and SASP can contribute to the pathogenesis of periodontal diseases as well. In this study, we investigated the effect of DQ interventions on periodontal tissue health using preclinical models of aging. In vitro, DQ ameliorated biological signatures of senescence in human gingival keratinocytes upon persistent exposure to periodontal bacteria, Fusobacterium nucleatum, by modulating the levels of key senescence markers such as p16, SA-β-galactosidase, and lamin-B1 and inflammatory mediators associated with SASP including interleukin-8, matrix metalloproteinase (MMP)-1, and MMP-3. In vivo, the oral administration of DQ mitigated senescent cell burden and SASP in gingival tissues and reduced naturally progressing periodontal bone loss in aged mice. Collectively, our findings provide proof-of-concept evidence for translational studies and reveal that targeting gingival senescence and the senescence-associated secretome can be an effective strategy to improve periodontal health, particularly in vulnerable populations.
Collapse
Affiliation(s)
- K. Rattanaprukskul
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - X.-J. Xia
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M. Hysa
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M. Jiang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M. Hung
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S.F. Suslavich
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S.E. Sahingur
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
61
|
Czajkowski K, Herbet M, Murias M, Piątkowska-Chmiel I. Senolytics: charting a new course or enhancing existing anti-tumor therapies? Cell Oncol (Dordr) 2025; 48:351-371. [PMID: 39633108 PMCID: PMC11996976 DOI: 10.1007/s13402-024-01018-5] [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] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Cell senescence is a natural response within our organisms. Initially, it was considered an effective anti-tumor mechanism. However, it is now believed that while cell senescence initially acts as a robust barrier against tumor initiation, the subsequent accumulation of senescent cells can paradoxically promote cancer recurrence and cause damage to neighboring tissues. This intricate balance between cell proliferation and senescence plays a pivotal role in maintaining tissue homeostasis. Moreover, senescence cells secrete many bioactive molecules collectively termed the senescence-associated secretory phenotype (SASP), which can induce chronic inflammation, alter tissue architecture, and promote tumorigenesis through paracrine signaling. Among the myriads of compounds, senotherapeutic drugs have emerged as exceptionally promising candidates in anticancer treatment. Their ability to selectively target senescent cells while sparing healthy tissues represents a paradigm shift in therapeutic intervention, offering new avenues for personalized oncology medicine. Senolytics have introduced new therapeutic possibilities by enabling the targeted removal of senescent cells. As standalone agents, they can clear tumor cells in a senescent state and, when combined with chemo- or radiotherapy, eliminate residual senescent cancer cells after treatment. This dual approach allows for the intentional use of lower-dose therapies or the removal of unintended senescent cells post-treatment. Additionally, by targeting non-cancerous senescent cells, senolytics may help reduce tumor formation risk, limit recurrence, and slow disease progression. This article examines the mechanisms of cellular senescence, its role in cancer treatment, and the importance of senotherapy, with particular attention to the therapeutic potential of senolytic drugs.
Collapse
Affiliation(s)
- Konrad Czajkowski
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Mariola Herbet
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, Poznań, Poland
| | - Iwona Piątkowska-Chmiel
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
62
|
Olascoaga S, Konigsberg M, Espinal‐Enríquez J, Tovar H, Matadamas‐Martínez F, Pérez‐Villanueva J, López‐Diazguerrero NE. Transcriptomic signatures and network-based methods uncover new senescent cell anti-apoptotic pathways and senolytics. FEBS J 2025; 292:1950-1971. [PMID: 39871113 PMCID: PMC12001159 DOI: 10.1111/febs.17402] [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: 06/11/2024] [Revised: 09/27/2024] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Cellular senescence is an irreversible cell cycle arrest caused by various stressors that damage cells. Over time, senescent cells accumulate and contribute to the progression of multiple age-related degenerative diseases. It is believed that these cells accumulate partly due to their ability to evade programmed cell death through the development and activation of survival and antiapoptotic resistance mechanisms; however, many aspects of how these survival mechanisms develop and activate are still unknown. By analyzing transcriptomic signature profiles generated by the LINCS L1000 project and using network-based methods, we identified various genes that could represent new senescence-related survival mechanisms. Additionally, employing the same methodology, we identified over 600 molecules with potential senolytic activity. Experimental validation of our computational findings confirmed the senolytic activity of Fluorouracil, whose activity would be mediated by a multitarget mechanism, revealing that its targets AURKA, EGFR, IRS1, SMAD4, and KRAS are new senescent cell antiapoptotic pathways (SCAPs). The development of these pathways could depend on the stimulus that induces cellular senescence. The SCAP development and activation mechanisms proposed in this work offer new insights into how senescent cells survive. Identifying new antiapoptotic resistance targets and drugs with potential senolytic activity paves the way for developing new pharmacological therapies to eliminate senescent cells selectively.
Collapse
Affiliation(s)
- Samael Olascoaga
- Posgrado en Biología Experimental, DCBSUniversidad Autónoma Metropolitana IztapalapaMexico CityMexico
- Laboratorio de Bioenergética y Envejecimiento Celular, Departamento de Ciencias de la SaludUniversidad Autónoma Metropolitana‐IztapalapaMexico CityMexico
| | - Mina Konigsberg
- Laboratorio de Bioenergética y Envejecimiento Celular, Departamento de Ciencias de la SaludUniversidad Autónoma Metropolitana‐IztapalapaMexico CityMexico
| | | | - Hugo Tovar
- Computational Genomics DivisionNational Institute of Genomic MedicineMexico CityMexico
| | - Félix Matadamas‐Martínez
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias‐UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro SocialMexico CityMexico
| | - Jaime Pérez‐Villanueva
- División de Ciencias Biológicas y de la Salud, Departamento de Sistemas BiológicosUniversidad Autónoma Metropolitana‐Xochimilco (UAM‐X)Mexico CityMexico
| | - Norma Edith López‐Diazguerrero
- Laboratorio de Bioenergética y Envejecimiento Celular, Departamento de Ciencias de la SaludUniversidad Autónoma Metropolitana‐IztapalapaMexico CityMexico
| |
Collapse
|
63
|
Bracken OV, De Maeyer RPH, Akbar AN. Enhancing immunity during ageing by targeting interactions within the tissue environment. Nat Rev Drug Discov 2025; 24:300-315. [PMID: 39875569 DOI: 10.1038/s41573-024-01126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/30/2025]
Abstract
Immunity declines with age. This results in a higher risk of age-related diseases, diminished ability to respond to new infections and reduced response to vaccines. The causes of this immune dysfunction are cellular senescence, which occurs in both lymphoid and non-lymphoid tissue, and chronic, low-grade inflammation known as 'inflammageing'. In this Review article, we highlight how the processes of inflammation and senescence drive each other, leading to loss of immune function. To break this cycle, therapies are needed that target the interactions between the altered tissue environment and the immune system instead of targeting each component alone. We discuss the relative merits and drawbacks of therapies that are directed at eliminating senescent cells (senolytics) and those that inhibit inflammation (senomorphics) in the context of tissue niches. Furthermore, we discuss therapeutic strategies designed to directly boost immune cell function and improve immune surveillance in tissues.
Collapse
Affiliation(s)
| | - Roel P H De Maeyer
- Division of Medicine, University College London, London, UK
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Arne N Akbar
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
64
|
Du K, Umbaugh DS, Wang L, Jun JH, Dutta RK, Oh SH, Ren N, Zhang Q, Ko DC, Ferreira A, Hill J, Gao G, Pullen SS, Jain V, Gregory S, Abdelmalek MF, Diehl AM. Targeting senescent hepatocytes for treatment of metabolic dysfunction-associated steatotic liver disease and multi-organ dysfunction. Nat Commun 2025; 16:3038. [PMID: 40155379 PMCID: PMC11953480 DOI: 10.1038/s41467-025-57616-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/23/2025] [Indexed: 04/01/2025] Open
Abstract
Senescent hepatocytes accumulate in metabolic dysfunction-associated steatotic liver disease (MASLD) and are linked to worse clinical outcomes. However, their heterogeneity and lack of specific markers have made them difficult to target therapeutically. Here, we define a senescent hepatocyte gene signature (SHGS) using in vitro and in vivo models and show that it tracks with MASLD progression/regression across mouse models and large human cohorts. Single-nucleus RNA-sequencing and functional studies reveal that SHGS+ hepatocytes originate from p21+ cells, lose key liver functions and release factors that drive disease progression. One such factor, GDF15, increases in circulation alongside SHGS+ burden and disease progression. Through chemical screening, we identify senolytics that selectively eliminate SHGS+ hepatocytes and improve MASLD in male mice. Notably, SHGS enrichment also correlates with dysfunction in other organs. These findings establish SHGS+ hepatocytes as key drivers of MASLD and highlight a potential therapeutic strategy for targeting senescent cells in liver disease and beyond.
Collapse
Affiliation(s)
- Kuo Du
- Department of Medicine, Duke University, Durham, NC, USA.
| | | | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Ji Hye Jun
- Department of Medicine, Duke University, Durham, NC, USA
| | - Rajesh K Dutta
- Department of Medicine, Duke University, Durham, NC, USA
| | - Seh Hoon Oh
- Department of Medicine, Duke University, Durham, NC, USA
| | - Niansheng Ren
- Department of Medicine, Duke University, Durham, NC, USA
| | - Qiaojuan Zhang
- Department of Neurology, Duke University, Durham, NC, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Ana Ferreira
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Jon Hill
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Guannan Gao
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Steven S Pullen
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Simon Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | | | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
65
|
Wang S, Huo T, Lu M, Zhao Y, Zhang J, He W, Chen H. Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies. Cells 2025; 14:499. [PMID: 40214453 PMCID: PMC11987807 DOI: 10.3390/cells14070499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Cellular senescence is an irreversible state of cell cycle arrest. Senescent cells (SCs) accumulate in the body with age and secrete harmful substances known as the senescence-associated secretory phenotype (SASP), causing chronic inflammation; at the same time, chronic inflammation leads to a decrease in immune system function, known as immunosenescence, which further accelerates the aging process. Cellular senescence and immunosenescence are closely related to a variety of chronic diseases, including cardiovascular diseases, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Studying the mechanisms of cellular senescence and immunosenescence and developing targeted interventions are crucial for improving the immune function and quality of life of elderly people. Here, we review a series of recent studies focusing on the molecular mechanisms of cellular senescence and immunosenescence, the regulation of aging by the immune system, and the latest advances in basic and clinical research on senolytics. We summarize the cellular and animal models related to aging research, as well as the mechanisms, strategies, and future directions of aging interventions from an immunological perspective, with the hope of laying the foundation for developing novel and practical anti-aging therapies.
Collapse
Affiliation(s)
- Shuaiqi Wang
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Tong Huo
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Mingyang Lu
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Yueqi Zhao
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Jianmin Zhang
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
| | - Wei He
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Hui Chen
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
| |
Collapse
|
66
|
Viteri JA, Kerr NR, Brennan CD, Kick GR, Wang M, Ketabforoush A, Snyder HK, Moore PJ, Darvishi FB, Dashtmian AR, Ayyagari SN, Rich K, Zhu Y, Arnold WD. Targeting senescence in Amyotrophic Lateral Sclerosis: senolytic treatment improves neuromuscular function and preserves cortical excitability in a TDP-43 Q331K mouse model. RESEARCH SQUARE 2025:rs.3.rs-6081213. [PMID: 40196013 PMCID: PMC11975006 DOI: 10.21203/rs.3.rs-6081213/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder marked by progressive motor neuron degeneration in the primary motor cortex (PMC) and spinal cord. Aging is a key factor in ALS onset and progression, with evidence suggesting that biological aging-a process involving cellular decline- far outpaces chronological aging in ALS. This promotes senescent cell accumulation-marked by irreversible cell-cycle arrest, impaired apoptosis, and chronic inflammation-disrupting tissue homeostasis and impairing neuronal support functions. Thus, targeting senescence presents a novel therapeutic strategy for ALS. Here, we investigated the senolytic combination Dasatinib and Quercetin (D&Q) in TDP-43Q331K ALS mice. D&Q improved neuromuscular function and reduced plasma neurofilament light chain, a biomarker of axonal damage. The most pronounced improvement was the improved cortical excitability, accompanied by reductions in senescence and TDP-43 in the PMC. These findings highlight the potential of senolytics to mitigate ALS-related dysfunction, supporting their viability as a therapeutic strategy.
Collapse
Affiliation(s)
- Jose A Viteri
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Nathan R Kerr
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Charles D Brennan
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Grace R Kick
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, MO USA
| | - Meifang Wang
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Arsh Ketabforoush
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Harper K Snyder
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Peter J Moore
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Fereshteh B Darvishi
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Anna Roshani Dashtmian
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Sindhuja N Ayyagari
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| | - Kelly Rich
- Department of Genetics, Harvard Medical School, Boston, MA USA
| | - Yi Zhu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; Robert & Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - W David Arnold
- Department of Physical Medicine and Rehabilitation, University of Missouri-Columbia, Columbia, MO USA; NextGen Precision Health, University of Missouri-Columbia, Columbia, MO USA
| |
Collapse
|
67
|
Chen L, Wu B, Mo L, Chen H, Yin X, Zhao Y, Cui Z, Cui F, Chen L, Deng Q, Gao C, Yao P, Li Y, Tang Y. High-content screening identifies ganoderic acid A as a senotherapeutic to prevent cellular senescence and extend healthspan in preclinical models. Nat Commun 2025; 16:2878. [PMID: 40128218 PMCID: PMC11933296 DOI: 10.1038/s41467-025-58188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Accumulated senescent cells during the aging process are a key driver of functional decline and age-related disorders. Here, we identify ganoderic acid A (GAA) as a potent anti-senescent compound with low toxicity and favorable drug properties through high-content screening. GAA, a major natural component of Ganoderma lucidum, possesses broad-spectrum geroprotective activity across various species. In C. elegans, GAA treatment extends lifespan and healthspan as effectively as rapamycin. Administration of GAA also mitigates the accumulation of senescent cells and physiological decline in multiple organs of irradiation-stimulated premature aging mice, natural aged mice, and western diet-induced obese mice. Notably, GAA displays a capability to enhance physical function and adapts to conditional changes in metabolic demand as mice aged. Mechanistically, GAA directly binds to TCOF1 to maintain ribosome homeostasis and thereby alleviate cellular senescence. These findings suggest a feasible senotherapeutic strategy for protecting against cellular senescence and age-related pathologies.
Collapse
Affiliation(s)
- Li Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Wuhan, Hubei, China
| | - Bangfu Wu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Mo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huimin Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingzhu Yin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - ZhaoYu Cui
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feipeng Cui
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianchun Deng
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Wuhan, Hubei, China
| | - Chao Gao
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanyan Li
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
68
|
Bar S, Hilsabeck TA, Pattavina B, López-Domínguez JA, Basisty N, Bons J, Watson M, Schilling B, Campisi J, Kapahi P, Sharma A. Inhibition of the metalloprotease ADAM19 as a novel senomorphic strategy to ameliorate gut permeability and senescence markers by modulating senescence-associated secretory phenotype (SASP). Aging (Albany NY) 2025; 17:757-777. [PMID: 40117561 PMCID: PMC11984429 DOI: 10.18632/aging.206224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 03/06/2025] [Indexed: 03/23/2025]
Abstract
Accumulation of DNA damage can accelerate aging through cellular senescence. Previously, we established a Drosophila model to investigate the effects of radiation-induced DNA damage on the intestine. In this model, we examined irradiation-responsive senescence in the fly intestine. Through an unbiased genome-wide association study (GWAS) utilizing 156 strains from the Drosophila Genetic Reference Panel (DGRP), we identified meltrin (the drosophila orthologue of mammalian ADAM19) as a potential modulator of the senescence-associated secretory phenotype (SASP). Knockdown of meltrin resulted in reduced gut permeability, DNA damage, and expression of the senescence marker β-galactosidase (SA-β-gal) in the fly gut following irradiation. Additionally, inhibition of ADAM19 in mice using batimastat-94 reduced gut permeability and inflammation in the gut. Our findings extend to human primary fibroblasts, where ADAM19 knockdown or pharmacological inhibition decreased expression of specific SASP factors and SA-β-gal. Furthermore, proteomics analysis of the secretory factor of senescent cells revealed a significant decrease in SASP factors associated with the ADAM19 cleavage site. These data suggest that ADAM19 inhibition could represent a novel senomorphic strategy.
Collapse
Affiliation(s)
- Sudipta Bar
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Blaine Pattavina
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Jackson Laboratory, Farmington, CT 06032, USA
| | - José Alberto López-Domínguez
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Instituto de Biología Molecular y Celular del Cáncer and Centro de Investigación del Cáncer of Salamanca, University of Salamanca-CSIC, Campus Unamuno s/n, 37007 Salamanca, Spain
| | - Nathan Basisty
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Mark Watson
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Amit Sharma
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- SENS Research Foundation, Mountain View, CA 94041, USA
| |
Collapse
|
69
|
Zhang X, Gao Y, Zhang S, Wang Y, Du Y, Hao S, Ni T. The Regulation of Cellular Senescence in Cancer. Biomolecules 2025; 15:448. [PMID: 40149983 PMCID: PMC11940315 DOI: 10.3390/biom15030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Cellular senescence is a stable state of cell cycle arrest caused by telomere shortening or various stresses. After senescence, cells cease dividing and exhibit many age-related characteristics. Unlike the halted proliferation of senescence cells, cancer cells are considered to have unlimited growth potential. When cells display senescence-related features, such as telomere loss or stem cell failure, they can inhibit tumor development. Therefore, inducing cells to enter a senescence state can serve as a barrier to tumor cell development. However, many recent studies have found that sustained senescence of tumor cells or normal cells under certain circumstances can exert environment-dependent effects of tumor promotion and inhibition by producing various cytokines. In this review, we first introduce the causes and characteristics of induced cellular senescence, analyze the senescence process of immune cells and cancer cells, and then discuss the dual regulatory role of cell senescence on tumor growth and senescence-induced therapies targeting cancer cells. Finally, we discuss the role of senescence in tumor progression and treatment opportunities, and propose further studies on cellular senescence and cancer therapy.
Collapse
Affiliation(s)
- Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Siyu Zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China;
| | - Yixiong Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| |
Collapse
|
70
|
Mannarino M, Cherif H, Ghazizadeh S, Martinez OW, Sheng K, Cousineau E, Lee S, Millecamps M, Gao C, Gilbert A, Peirs C, Naeini RS, Ouellet JA, S. Stone L, Haglund L. Senolytic treatment for low back pain. SCIENCE ADVANCES 2025; 11:eadr1719. [PMID: 40085710 PMCID: PMC11908501 DOI: 10.1126/sciadv.adr1719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025]
Abstract
Senescent cells (SnCs) accumulate because of aging and external cellular stress throughout the body. They adopt a senescence-associated secretory phenotype (SASP) and release inflammatory and degenerative factors that actively contribute to age-related diseases, such as low back pain (LBP). The senolytics, o-vanillin and RG-7112, remove SnCs in human intervertebral discs (IVDs) and reduce SASP release, but it is unknown whether they can treat LBP. sparc-/- mice, with LBP, were treated orally with o-vanillin and RG-7112 as single or combination treatments. Treatment reduced LBP and SASP factor release and removed SnCs from the IVD and spinal cord. Treatment also lowered degeneration scores in the IVDs, improved vertebral bone quality, and reduced the expression of pain markers in the spinal cord. Together, our data suggest RG-7112 and o-vanillin as potential disease-modifying drugs for LBP and other painful disorders linked to cell senescence.
Collapse
Affiliation(s)
- Matthew Mannarino
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- ABC-platform (Animal Behavioral Characterization) at the Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
| | - Hosni Cherif
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, QC, Canada
| | - Saber Ghazizadeh
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
| | - Oliver Wu Martinez
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
| | - Kai Sheng
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Shriner’s Hospital for Children, Montreal, QC, Canada
| | - Elsa Cousineau
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
| | - Seunghwan Lee
- ABC-platform (Animal Behavioral Characterization) at the Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN, USA
| | - Magali Millecamps
- ABC-platform (Animal Behavioral Characterization) at the Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
| | - Chan Gao
- Division of Physiatry, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Alice Gilbert
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Physiology and Cell Information Systems, McGill University, Montreal, QC, Canada
- Université Clermont-Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, Clermont-Ferrand, France
| | - Cedric Peirs
- Université Clermont-Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, Clermont-Ferrand, France
| | - Reza Sharif Naeini
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Physiology and Cell Information Systems, McGill University, Montreal, QC, Canada
| | - Jean A. Ouellet
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, QC, Canada
- Shriner’s Hospital for Children, Montreal, QC, Canada
| | - Laura S. Stone
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN, USA
| | - Lisbet Haglund
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, QC, Canada
- Shriner’s Hospital for Children, Montreal, QC, Canada
| |
Collapse
|
71
|
Chen Y, Jiang F, Zeng Y, Zhang M. The role of retinal pigment epithelial senescence and the potential of senotherapeutics in age-related macular degeneration. Surv Ophthalmol 2025:S0039-6257(25)00053-0. [PMID: 40089029 DOI: 10.1016/j.survophthal.2025.03.004] [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: 10/14/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment in the aging population. Evidence showing the presence of cellular senescence in retinal pigment epithelium (RPE) of patients with AMD is growing. Senescent RPE play a pivotal role in its pathogenesis. The senescent RPE suffers from structural and functional alterations and disruption of the surrounding microenvironment due to the development of the senescence-associated secretory phenotype, which contributes to metabolic dysfunctions and inflammatory responses in the retina. Senotherapeutics, including senolytics, senomorphics and others, are novel treatments targeting senescent cells and are promising treatments for AMD. As senotherapeutic targets are being developed, it is promising that the burden of AMD could be decreased.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Feipeng Jiang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Yue Zeng
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Meixia Zhang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| |
Collapse
|
72
|
Sanz-Ros J, Huete-Acevedo J, Mas-Bargues C, Romero-García N, Dromant M, van Weeghel M, Janssens GE, Borrás C. Small extracellular vesicles from young adipose-derived stem cells ameliorate age-related changes in the heart of old mice. Stem Cell Res Ther 2025; 16:138. [PMID: 40082997 PMCID: PMC11907833 DOI: 10.1186/s13287-025-04255-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Aging entails a progressive decline in physiological functions, elevating the risk of age-related diseases like heart failure or aortic stenosis. Stem cell therapies, especially those that use paracrine signaling, can potentially mitigate the adverse effects of aging. OBJECTIVES The objective is to explore the potential of small extracellular vesicles (sEVs) derived from young adipose-derived stem cells (ADSC-sEVs) in reversing structural, molecular, and functional changes associated with aging in the heart. METHODS Aged C57BL/6J mice were treated intravenously with ADSC-sEVs from young mice or PBS as controls. Young mice were included to identify specific age-associated changes. The impact of sEV treatment on cardiac function was assessed using transthoracic echocardiography and physical endurance tests. Histological and molecular analyses were conducted on heart tissue to evaluate structural changes and markers of senescence, inflammation, and oxidative stress. A comprehensive metabolomic analysis was also performed on heart tissues to identify changes in metabolic profiles associated with aging and treatment status. RESULTS The administration of ADSC-sEVs significantly improves several aging-associated cardiac parameters, including oxidative stress, inflammation, and cellular senescence reductions. We also report on the age-related reversal of myocardial structure and function changes, highlighted by decreased fibrosis and improved vascularization. Notably, echocardiographic assessments reveal that sEV treatments ameliorate diastolic dysfunction and left ventricle structural alterations typically associated with aging. Furthermore, the treatment shifts the heart metabolome towards a more youthful profile. CONCLUSIONS These results denote the potential of ADSC-sEVs as a novel, noninvasive therapeutic strategy for mitigating cardiac aging-associated functional decline.
Collapse
Affiliation(s)
- Jorge Sanz-Ros
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia, Spain
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Javier Huete-Acevedo
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia, Spain
| | - Cristina Mas-Bargues
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia, Spain
| | - Nekane Romero-García
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia, Spain
- Department of Anesthesiology and Surgical Trauma Intensive Care, Hospital Clinic Universitari Valencia, University of Valencia, Valencia, 46010, Spain
| | - Mar Dromant
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia, Spain
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam Gastroenterology, Endocrinology, and Metabolism, University of Amsterdam, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam Gastroenterology, Endocrinology, and Metabolism, University of Amsterdam, Amsterdam, The Netherlands
| | - Consuelo Borrás
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia, Spain.
| |
Collapse
|
73
|
Rahbar Saadat Y, Abbasi A, Hejazian SS, Hekmatshoar Y, Ardalan M, Farnood F, Zununi Vahed S. Combating chronic kidney disease-associated cachexia: A literature review of recent therapeutic approaches. BMC Nephrol 2025; 26:133. [PMID: 40069669 PMCID: PMC11895341 DOI: 10.1186/s12882-025-04057-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/05/2025] [Indexed: 03/15/2025] Open
Abstract
In 2008, the Society on Sarcopenia, Cachexia, and Wasting Disorders introduced a generic definition for all types of cachexia: "a complex metabolic syndrome associated with the underlying illness characterized by a loss of muscle, with or without fat loss". It is well-known that the presence of inflammatory burden in end-stage renal disease (ESRD) patients may lead to the evolution of cachexia. Since the etiology of cachexia in chronic kidney disease (CKD) is multifactorial, thus the successful treatment must involve several concomitant measures (nutritional interventions, appetite stimulants, and anti-inflammatory pharmacologic agents) to provide integrated effective therapeutic modalities to combat causative factors and alleviate the outcomes of patients. Given the high mortality rate associated with cachexia, developing new therapeutic modalities are prerequisite for ameliorating patients with CKD worldwide. The present review aims to discuss some therapeutic strategies and provide an update on advances in nutritional approaches to counteract cachexia.
Collapse
Affiliation(s)
| | - Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyyed Sina Hejazian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Hekmatshoar
- Medical Biology Department, School of Medicine, Altinbas University, Istanbul, Türkiye
| | | | - Farahnoosh Farnood
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
74
|
Wang L, Tang D. Immunosenescence promotes cancer development: from mechanisms to treatment strategies. Cell Commun Signal 2025; 23:128. [PMID: 40065335 PMCID: PMC11892258 DOI: 10.1186/s12964-025-02082-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
The body's innate immune system plays a pivotal role in identifying and eliminating cancer cells. However, as the immune system ages, its functionality can deteriorate, becoming dysfunctional, inefficient, or even inactive-a condition referred to as immunosenescence. This decline significantly increases the risk of malignancies. While the pro-cancer effects of T-cell aging have been widely explored, there remains a notable gap in the literature regarding the impact of aging on innate immune cells, such as macrophages and neutrophils. This review seeks to address this gap, with emphasis on these cell types. Furthermore, although certain cancer immunotherapies, including immune checkpoint inhibitors (ICIs), have demonstrated efficacy across a broad spectrum of cancers, elderly patients are less likely to derive clinical benefit from these treatments. In some cases, they may even experience immune-related adverse events (irAEs). While senolytic strategies have shown promise in exerting anti-cancer effects, their adverse reactions and potential off-target effects present significant challenges. This review aims to elucidate the pro-cancer effects of immunosenescence, its implications for the efficacy and safety of ICIs, and potential anti-aging treatment strategies. In addition, optimizing anti-aging therapies to minimize adverse reactions and enhance therapeutic outcomes remains a critical focus for future research endeavors.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University; Northern Jiangsu People's Hospital; The Yangzhou Clinical Medical College of Xuzhou Medical University; The Yangzhou School of Clinical Medicine of Dalian Medical University; The Yangzhou School of Clinical Medicine of Nanjing Medical University; Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225000, China.
| |
Collapse
|
75
|
Gross PS, Durán-Laforet V, Ho LT, Melchor GS, Zia S, Manavi Z, Barclay WE, Lee SH, Shults N, Selva S, Alvarez E, Plemel JR, Fu MM, Schafer DP, Huang JK. Senescent-like microglia limit remyelination through the senescence associated secretory phenotype. Nat Commun 2025; 16:2283. [PMID: 40055369 PMCID: PMC11889183 DOI: 10.1038/s41467-025-57632-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
The capacity to regenerate myelin in the central nervous system diminishes with age. This decline is particularly evident in multiple sclerosis (MS), a chronic demyelinating disease. Whether cellular senescence, a hallmark of aging, contributes to remyelination impairment remains unknown. Here, we show that senescent cells accumulate within demyelinated lesions after injury, and treatments with senolytics enhances remyelination in young and middle-aged mice but not aged mice. In young mice, we observe the upregulation of senescence-associated transcripts, primarily in microglia and macrophages, after demyelination, followed by a reduction during remyelination. However, in aged mice, senescence-associated factors persist within lesions, correlating with inefficient remyelination. Proteomic analysis of the senescence-associated secretory phenotype (SASP) reveals elevated levels of CCL11/Eotaxin-1 in lesions of aged mice, which is found to inhibit oligodendrocyte maturation. These results suggest therapeutic targeting of SASP components, such as CCL11, may improve remyelination in aging and MS.
Collapse
Affiliation(s)
- Phillip S Gross
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Violeta Durán-Laforet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lana T Ho
- Department of Molecular and Cell Biology (MCB), University of California Berkeley, Berkeley, CA, USA
| | - George S Melchor
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Zeeba Manavi
- Department of Biology, Georgetown University, Washington, DC, USA
| | - William E Barclay
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sung Hyun Lee
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Nataliia Shults
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Sean Selva
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Enrique Alvarez
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Meng-Meng Fu
- Department of Molecular and Cell Biology (MCB), University of California Berkeley, Berkeley, CA, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeffrey K Huang
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA.
- Department of Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
76
|
Nie T, Nepovimova E, Wu Q. Circadian rhythm, hypoxia, and cellular senescence: From molecular mechanisms to targeted strategies. Eur J Pharmacol 2025; 990:177290. [PMID: 39863143 DOI: 10.1016/j.ejphar.2025.177290] [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: 10/25/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Cellular senescence precipitates a decline in physiological activities and metabolic functions, often accompanied by heightened inflammatory responses, diminished immune function, and impaired tissue and organ performance. Despite extensive research, the mechanisms underpinning cellular senescence remain incompletely elucidated. Emerging evidence implicates circadian rhythm and hypoxia as pivotal factors in cellular senescence. Circadian proteins are central to the molecular mechanism governing circadian rhythm, which regulates homeostasis throughout the body. These proteins mediate responses to hypoxic stress and influence the progression of cellular senescence, with protein Brain and muscle arnt-like 1 (BMAL1 or Arntl) playing a prominent role. Hypoxia-inducible factor-1α (HIF-1α), a key regulator of oxygen homeostasis within the cellular microenvironment, orchestrates the transcription of genes involved in various physiological processes. HIF-1α not only impacts normal circadian rhythm functions but also can induce or inhibit cellular senescence. Notably, HIF-1α may aberrantly interact with BMAL1, forming the HIF-1α-BMAL1 heterodimer, which can instigate multiple physiological dysfunctions. This heterodimer is hypothesized to modulate cellular senescence by affecting the molecular mechanism of circadian rhythm and hypoxia signaling pathways. In this review, we elucidate the intricate relationships among circadian rhythm, hypoxia, and cellular senescence. We synthesize diverse evidence to discuss their underlying mechanisms and identify novel therapeutic targets to address cellular senescence. Additionally, we discuss current challenges and suggest potential directions for future research. This work aims to deepen our understanding of the interplay between circadian rhythm, hypoxia, and cellular senescence, ultimately facilitating the development of therapeutic strategies for aging and related diseases.
Collapse
Affiliation(s)
- Tong Nie
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
77
|
Suda M, Tchkonia T, Kirkland JL, Minamino T. Targeting senescent cells for the treatment of age-associated diseases. J Biochem 2025; 177:177-187. [PMID: 39727337 DOI: 10.1093/jb/mvae091] [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: 10/15/2024] [Revised: 11/18/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
Cellular senescence, which entails cellular dysfunction and inflammatory factor release-the senescence-associated secretory phenotype (SASP)-is a key contributor to multiple disorders, diseases and the geriatric syndromes. Targeting senescent cells using senolytics has emerged as a promising therapeutic strategy for these conditions. Among senolytics, the combination of dasatinib and quercetin (D + Q) was the earliest and one of the most successful so far. D + Q delays, prevents, alleviates or treats multiple senescence-associated diseases and disorders with improvements in healthspan across various pre-clinical models. While early senolytic therapies have demonstrated promise, ongoing research is crucial to refine them and address such challenges as off-target effects. Recent advances in senolytics include new drugs and therapies that target senescent cells more effectively. The identification of senescence-associated antigens-cell surface molecules on senescent cells-pointed to another promising means for developing novel therapies and identifying biomarkers of senescent cell abundance.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo City, Tokyo 113-8431, Japan
- Division of Endocrinology, Diabetes, & Metabolism, Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, 8687 Melrose Ave, Pacific Design Center, West Hollywood, CA 90069, USA
| | - Tamar Tchkonia
- Division of Endocrinology, Diabetes, & Metabolism, Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, 8687 Melrose Ave, Pacific Design Center, West Hollywood, CA 90069, USA
| | - James L Kirkland
- Division of Endocrinology, Diabetes, & Metabolism, Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, 8687 Melrose Ave, Pacific Design Center, West Hollywood, CA 90069, USA
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo City, Tokyo 113-8431, 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, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
78
|
Meguro S, Nakanishi M. Cellular senescence in the cancer microenvironment. J Biochem 2025; 177:171-176. [PMID: 39760850 DOI: 10.1093/jb/mvaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 01/07/2025] Open
Abstract
In this ageing society, the number of patients suffering from age-related diseases, including cancer, is increasing. Cellular senescence is a cell fate that involves permanent cell cycle arrest. Accumulated senescent cells in tissues over time present senescence-associated secretory phenotype (SASP) and make the inflammatory context, disturbing the tumour microenvironment. In particular, the effect of senescent cancer-associated fibroblasts on cancer progression has recently come under the spotlight. Although scientific evidence on the impact of cellular senescence on cancer is emerging, the association between cellular senescence and cancer is heterogeneous and the comprehensive mechanism is still not revealed. Recently, a therapy targeting senescent cells, senotherapeutics, has been reported to be effective against cancer in preclinical research and even clinical trials. With further research, the development of senotherapeutics as a novel cancer therapy is expected.
Collapse
Affiliation(s)
- Satoru Meguro
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Tokyo, Japan
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1247, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Tokyo, Japan
| |
Collapse
|
79
|
Foster TC, Kumar A. Sex, senescence, senolytics, and cognition. Front Aging Neurosci 2025; 17:1555872. [PMID: 40103928 PMCID: PMC11913825 DOI: 10.3389/fnagi.2025.1555872] [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: 01/05/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
This review focuses on sexual dimorphism in cellular senescence and senolytic treatment in relation to brain health and age-related cognitive decline. The stressors of aging, DNA damage, inflammation, and oxidative stress induce cell senescence, a hallmark of aging. Senescent cells change their function and molecular profile and are primed to release pro-inflammatory cytokines. The functional changes include the activation of cell signals to prevent cell death. The release of pro-inflammatory cytokines from peripheral senescent cells during middle age induces senescence of neighbor cells and heightens the level of systemic inflammation, contributing to neuroinflammation. In response to neuroinflammation and oxidative stress, some neurons alter their physiology, decreasing neuronal excitability and synaptic transmission. Senescent neurophysiology is protective against cell death due to excitotoxicity, at the expense of a loss of normal cell function, contributing to age-related cognitive decline. The level of peripheral cell senescence and systemic inflammation may underlie sexual dimorphism in the prevalence, symptoms, and pathogenesis of age-related diseases, including neurodegenerative diseases. Sex differences have been observed for senescence of astrocytes, microglia, and peripheral cells, including those involved in innate and adaptive immune responses. Interventions that remove senescent cells, such as senolytic drugs, can reduce or ameliorate some of the aging-related loss of function. Similarities and differences in senolytic responses of males and females depend on the system examined, the treatment regimen, the level of senescent cell burden, and the age when treatment is initiated. Estrogen impacts several of these factors and influences the transcription of genes promoting growth, proliferation, and cell survival programs in a manner opposite that of senolytic drugs. In addition, estrogen has anti-aging effects that are independent of cell senescence, including rapidly modifying senescent neurophysiology. Thus, it is important to recognize that, in addition to sex differences in cell senescence, there are other sexually dimorphic mechanisms that contribute to the aging process. The results indicate that senolytics interact with fundamental biology, including sex hormones.
Collapse
Affiliation(s)
- Thomas C Foster
- McKnight Brain Institute, Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Genetics and Genomics Graduate Program, Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Ashok Kumar
- McKnight Brain Institute, Department of Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
80
|
Nakano Y, Johmura Y. Functional diversity of senescent cells in driving ageing phenotypes and facilitating tissue regeneration. J Biochem 2025; 177:189-195. [PMID: 39760855 DOI: 10.1093/jb/mvae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/07/2025] Open
Abstract
As the global population continues to age, understanding the complex role of cellular senescence and its implications in healthy lifespans has gained increasing prominence. Cellular senescence is defined as the irreversible cessation of cell proliferation, accompanied by the secretion of a range of pro-inflammatory factors, collectively termed the senescence-associated secretory phenotype (SASP), in response to various cellular stresses. While the accumulation of senescent cells has been strongly implicated in the ageing process and the pathogenesis of age-related diseases owing to their pro-inflammatory properties, recent research has also highlighted their essential roles in processes such as tumour suppression, tissue development and repair. This review provides a comprehensive examination of the dual nature of senescent cells, evaluating their deleterious contributions to chronic inflammation, tissue dysfunction and disease, as well as their beneficial roles in maintaining physiological homeostasis. Additionally, we explored the therapeutic potential of senolytic agents designed to selectively eliminate detrimental senescent cells while considering the delicate balance between transient and beneficial senescence and the persistence of pathological senescence. A deeper understanding of these dynamics is critical to develop novel interventions aimed at mitigating age-related dysfunctions and enhancing healthy life expectancies.
Collapse
Affiliation(s)
- Yasuhiro Nakano
- Division of Cancer and Senescence Biology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192 Japan
- Integrated Systems of Aging Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192 Japan
| | - Yoshikazu Johmura
- Division of Cancer and Senescence Biology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192 Japan
- Integrated Systems of Aging Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192 Japan
| |
Collapse
|
81
|
Alshaebi F, Sciortino A, Kayed R. The Role of Glial Cell Senescence in Alzheimer's Disease. J Neurochem 2025; 169:e70051. [PMID: 40130281 PMCID: PMC11934031 DOI: 10.1111/jnc.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/26/2025]
Abstract
Glial cell senescence, characterized by the irreversible arrest of cell division and a pro-inflammatory secretory phenotype, has emerged as a critical player in the pathogenesis of Alzheimer's disease (ad). While much attention has been devoted to the role of neurons in ad, growing evidence suggests that glial cells, including astrocytes, microglia, and oligodendrocytes, contribute significantly to disease progression through senescence. In this review, we explore the molecular mechanisms underlying glial cell senescence in ad, focusing on the cellular signaling pathways, including DNA damage response and the accumulation of senescence-associated secretory phenotypes (SASP). We also examine how senescent glial cells exacerbate neuroinflammation, disrupt synaptic function, and promote neuronal death in ad. Moreover, we discuss emerging therapeutic strategies aimed at targeting glial cell senescence to mitigate the neurodegenerative processes in ad. By providing a comprehensive overview of current research on glial cell senescence in Alzheimer's disease, this review highlights its potential as a novel therapeutic target in the fight against ad.
Collapse
Affiliation(s)
- Fadhl Alshaebi
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Alessia Sciortino
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
82
|
Haider S, Sassu E, Stefanovska D, Stoyek MR, Preissl S, Hortells L. News from the old: Aging features in the intracardiac, musculoskeletal, and enteric nervous systems. Ageing Res Rev 2025; 105:102690. [PMID: 39947485 DOI: 10.1016/j.arr.2025.102690] [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: 07/15/2024] [Revised: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Aging strongly affects the peripheral nervous system (PNS), triggering alterations that vary depending on the innervated tissue. The most frequent alteration in peripheral nerve aging is reduced nerve fiber and glial density which can lead to abnormal nerve functionality. Interestingly, the activation of a destructive phenotype takes place in macrophages across the PNS while a reduced number of neuronal bodies is a unique feature of some enteric ganglia. Single cell/nucleus RNA-sequencing has unveiled a striking complexity of cell populations in the peripheral nerves, and these refined cell type annotations could facilitate a better understanding of PNS aging. While the effects of senescence on individual PNS cell types requires further characterization, the use of senolytics appears to improve general PNS function in models of aging. Here, we review the current understanding of age-related changes of the intracardiac, musculoskeletal, and enteric nervous system sub-sections of the PNS, highlighting their commonalities and differences.
Collapse
Affiliation(s)
- Severin Haider
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
| | - Eliza Sassu
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
| | - Dragana Stefanovska
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
| | - Mathew R Stoyek
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany; Institute of Pharmaceutical Sciences, Pharmacology & Toxicology, University of Graz, Graz 8010, Austria; Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Luis Hortells
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany; Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Science, UiT-The Arctic University of Norway, Tromsø 9019, Norway.
| |
Collapse
|
83
|
Yang Y, Jn-Simon N, He Y, Sun C, Zhang P, Hu W, Tian T, Zeng H, Basha S, Huerta AS, Sun LZ, Yin XM, Hromas R, Zheng G, Pi L, Zhou D. A BCL-xL/BCL-2 PROTAC effectively clears senescent cells in the liver and reduces MASH-driven hepatocellular carcinoma in mice. NATURE AGING 2025; 5:386-400. [PMID: 39890936 DOI: 10.1038/s43587-025-00811-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/05/2024] [Indexed: 02/03/2025]
Abstract
Accumulation of senescent cells (SnCs) plays a causative role in many age-related diseases and has also been implicated in the pathogenesis and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Senolytics that can selectively kill SnCs have the potential to be developed as therapeutics for these diseases. Here we report the finding that 753b, a dual BCL-xL/BCL-2 proteolysis-targeting chimera (PROTAC), acts as a potent and liver-tropic senolytic. We found that treatment with 753b selectively reduced SnCs in the liver in aged mice and STAM mice in part due to its sequestration in the liver. Moreover, 753b treatment could effectively reduce the progression of MASLD and the development of hepatocellular carcinoma (HCC) in STAM mice even after the mice developed substantial metabolic dysfunction-associated steatohepatitis (MASH) and hepatic fibrosis. These findings suggest that BCL-xL/BCL-2 PROTACs have the potential to be developed as therapeutics for MASLD to reduce MASH-driven HCC.
Collapse
Affiliation(s)
- Yang Yang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Natacha Jn-Simon
- Department of Pathology, Tulane University, New Orleans, LA, USA
| | - Yonghan He
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Chunbao Sun
- Department of Pathology, Tulane University, New Orleans, LA, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Wanyi Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Tian Tian
- Department of Pathology, Tulane University, New Orleans, LA, USA
| | - Huadong Zeng
- Advanced Magnetic Resonance Imaging and Spectroscopy Facility, University of Florida, Gainesville, FL, USA
| | | | - Araceli S Huerta
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lu-Zhe Sun
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Xian-Ming Yin
- Department of Pathology, Tulane University, New Orleans, LA, USA
| | - Robert Hromas
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Liya Pi
- Department of Pathology, Tulane University, New Orleans, LA, USA.
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
84
|
Meyer M, Fourie C, van der Merwe H, Botha H, Engelbrecht AM. Targeting treatment resistance in cervical cancer: A new avenue for senolytic therapies. Adv Med Sci 2025; 70:33-43. [PMID: 39549742 DOI: 10.1016/j.advms.2024.11.001] [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: 04/05/2024] [Revised: 07/27/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024]
Abstract
Cervical cancer poses a significant global health challenge, particularly impacting women in economically developing nations. This disparity stems from a combination of factors, including inadequate screening infrastructure and resource limitations. However, the foremost contributor is the widespread lack of awareness and limited accessibility to Human Papillomavirus (HPV) vaccination, which is a key preventative measure against cervical cancer development. Despite advancements in cervical cancer prevention, treatment resistance remains a major hurdle in achieving improved patient outcomes. Cellular senescence, specifically the senescence-associated secretory phenotype (SASP) and its bidirectional relationship with the immune system, has been implicated in resistance to conventional cervical cancer chemotherapy treatments. The exact mechanisms by which this state of growth arrest and the associated changes in immune regulation contribute to cervical cancer progression and the associated drug resistance are not entirely understood. This underscores the necessity for innovative strategies to address the prevalence of treatment-resistant cervical cancer, with one promising avenue being the utilisation of senolytics. Senolytics are agents that have promising efficacy in clearing senescent cells from tumour tissues, however neither the utilisation of senolytics for addressing senescence-induced treatment resistance nor the potential integration of immunotherapy as senolytic agents in cervical cancer treatment has been explored to date. This review provides a concise overview of the mechanisms underlying senescence induction and the pivotal role of the immune system in this process. Additionally, it explores various senolytic approaches that hold significant potential for advancing cervical cancer research.
Collapse
Affiliation(s)
- Madré Meyer
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Carla Fourie
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Haynes van der Merwe
- Department of Obstetrics and Gynaecology, Stellenbosch University Medical Campus, Cape Town, South Africa
| | - Hennie Botha
- Department of Obstetrics and Gynaecology, Stellenbosch University Medical Campus, Cape Town, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
85
|
Légaré C, Berglund JA, Duchesne E, Dumont NA. New Horizons in Myotonic Dystrophy Type 1: Cellular Senescence as a Therapeutic Target. Bioessays 2025; 47:e202400216. [PMID: 39723693 PMCID: PMC11848125 DOI: 10.1002/bies.202400216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Myotonic dystrophy type 1 (DM1) is considered a progeroid disease (i.e., causing premature aging). This hypervariable disease affects multiple systems, such as the musculoskeletal, central nervous, gastrointestinal, and others. Despite advances in understanding the underlying pathogenic mechanism of DM1, numerous gaps persist in our understanding, hindering elucidation of the heterogeneity and severity of its symptoms. Accumulating evidence indicates that the toxic intracellular RNA accumulation associated with DM1 triggers cellular senescence. These cells are in a state of irreversible cell cycle arrest and secrete a cocktail of cytokines, referred to as a senescence-associated secretory phenotype (SASP), that can have harmful effects on neighboring cells and more broadly. We hypothesize that cellular senescence contributes to the pathophysiology of DM1, and clearance of senescent cells is a promising therapeutic approach for DM1. We will discuss the therapeutic potential of different senotherapeutic drugs, especially senolytics that eliminate senescent cells, and senomorphics that reduce SASP expression.
Collapse
Affiliation(s)
- Cécilia Légaré
- RNA InstituteCollege of Arts and SciencesUniversity at Albany‐SUNYAlbanyNew YorkUSA
- School of Rehabilitation SciencesFaculty of MedicineUniversité LavalQuebecQuebecCanada
- CHU de Québec – Université Laval Research CenterQuébecQuébecCanada
- Groupe de Recherche Interdisciplinaire sur les Maladies Neuromusculaires (GRIMN)Centre intégré universitaire de santé et de services sociaux du Saguenay‐Lac‐Saint‐JeanSaguenayQuebecCanada
| | - J. Andrew Berglund
- RNA InstituteCollege of Arts and SciencesUniversity at Albany‐SUNYAlbanyNew YorkUSA
- Department of Biological Sciences, College of Arts and SciencesUniversity at Albany‐SUNYAlbanyNew YorkUSA
| | - Elise Duchesne
- School of Rehabilitation SciencesFaculty of MedicineUniversité LavalQuebecQuebecCanada
- CHU de Québec – Université Laval Research CenterQuébecQuébecCanada
- Groupe de Recherche Interdisciplinaire sur les Maladies Neuromusculaires (GRIMN)Centre intégré universitaire de santé et de services sociaux du Saguenay‐Lac‐Saint‐JeanSaguenayQuebecCanada
- Centre Interdisciplinaire de Recherche en Réadaptation et Intégration Sociale (Cirris)Centre Intégré Universitaire de Santé et de Services Sociaux Capitale‐NationaleQuébecQuebecCanada
| | - Nicolas A. Dumont
- CHU Sainte‐Justine Research CenterMontrealQuebecCanada
- School of rehabilitationFaculty of MedicineUniversité de MontréalMontrealQuebecCanada
| |
Collapse
|
86
|
Zhang M, Wei J, Sun Y, He C, Ma S, Pan X, Zhu X. The efferocytosis process in aging: Supporting evidence, mechanisms, and therapeutic prospects for age-related diseases. J Adv Res 2025; 69:31-49. [PMID: 38499245 PMCID: PMC11954809 DOI: 10.1016/j.jare.2024.03.008] [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: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Aging is characterized by an ongoing struggle between the buildup of damage caused by a combination of external and internal factors. Aging has different effects on phagocytes, including impaired efferocytosis. A deficiency in efferocytosis can cause chronic inflammation, aging, and several other clinical disorders. AIM OF REVIEW Our review underscores the possible feasibility and extensive scope of employing dual targets in various age-related diseases to reduce the occurrence and progression of age-related diseases, ultimately fostering healthy aging and increasing lifespan. Key scientific concepts of review Hence, the concurrent implementation of strategies aimed at augmenting efferocytic mechanisms and anti-aging treatments has the potential to serve as a potent intervention for extending the duration of a healthy lifespan. In this review, we comprehensively discuss the concept and physiological effects of efferocytosis. Subsequently, we investigated the association between efferocytosis and the hallmarks of aging. Finally, we discuss growing evidence regarding therapeutic interventions for age-related disorders, focusing on the physiological processes of aging and efferocytosis.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
87
|
Ma Y, Erb ML, Moore DJ. Aging, cellular senescence and Parkinson's disease. JOURNAL OF PARKINSON'S DISEASE 2025; 15:239-254. [PMID: 39973488 DOI: 10.1177/1877718x251316552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, affecting 1-2% of people over age 65. The risk of developing PD dramatically increases with advanced age, indicating that aging is likely a driving factor in PD neuropathogenesis. Several age-associated biological changes are also hallmarks of PD neuropathology, including mitochondrial dysfunction, oxidative stress, and neuroinflammation. Accumulation of senescent cells is an important feature of aging that contributes to age-related diseases. How age-related cellular senescence affects brain health and whether this phenomenon contributes to neuropathogenesis in PD is not yet fully understood. In this review, we highlight hallmarks of aging, including mitochondrial dysfunction, loss of proteostasis, genomic instability and telomere attrition in relation to well established PD neuropathological pathways. We then discuss the hallmarks of cellular senescence in the context of neuroscience and review studies that directly examine cellular senescence in PD. Studying senescence in PD presents challenges and holds promise for advancing our understanding of disease mechanisms, which could contribute to the development of effective disease-modifying therapeutics. Targeting senescent cells or modulating the senescence-associated secretory phenotype (SASP) in PD requires a comprehensive understanding of the complex relationship between PD pathogenesis and cellular senescence.
Collapse
Affiliation(s)
- Yue Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Madalynn L Erb
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
88
|
Thau H, Gerjol BP, Hahn K, von Gudenberg RW, Knoedler L, Stallcup K, Emmert MY, Buhl T, Wyles SP, Tchkonia T, Tullius SG, Iske J. Senescence as a molecular target in skin aging and disease. Ageing Res Rev 2025; 105:102686. [PMID: 39929368 DOI: 10.1016/j.arr.2025.102686] [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: 11/20/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Skin aging represents a multifactorial process influenced by both intrinsic and extrinsic factors, collectively known as the skin exposome. Cellular senescence, characterized by stable cell cycle arrest and secretion of pro-inflammatory molecules, has been implicated as a key driver of physiological and pathological skin aging. Increasing evidence points towards the role of senescence in a variety of dermatological diseases, where the accumulation of senescent cells in the epidermis and dermis exacerbates disease progression. Emerging therapeutic strategies such as senolytics and senomorphics offer promising avenues to target senescent cells and mitigate their deleterious effects, providing potential treatments for both skin aging and senescence-associated skin diseases. This review explores the molecular mechanisms of cellular senescence and its role in promoting age-related skin changes and pathologies, while compiling the observed effects of senotherapeutics in the skin and discussing the translational relevance.
Collapse
Affiliation(s)
- Henriette Thau
- Van Cleve Cardiac Regenerative Medicine Program Mayo Clinic, Rochester, Minesota, USA; Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bastian P Gerjol
- Department of Internal Medicine, Klinik Hirslanden, Zurich, Switzerland
| | - Katharina Hahn
- Department of Dermatology, Venereology and Allergology, Göttingen University Medical Center, Göttingen, Germany
| | - Rosalie Wolff von Gudenberg
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Leonard Knoedler
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany
| | - Kenneth Stallcup
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, Göttingen University Medical Center, Göttingen, Germany
| | | | - Tamar Tchkonia
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
89
|
Chen Y, Li J, Liu X, Geng Z, Xu K, Su J. Advances in biomarkers and diagnostic significance of organ aging. FUNDAMENTAL RESEARCH 2025; 5:683-696. [PMID: 40242549 PMCID: PMC11997494 DOI: 10.1016/j.fmre.2023.03.009] [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: 01/15/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 04/18/2025] Open
Abstract
A complete understanding of aging is a critical first step in treating age-related diseases and postponing aging dysfunction in the context of an aging global population. Aging in organisms is driven by related molecular alterations that gradually occur in many organs. There has previously been a wealth of knowledge of how cells behave as they age, but when aging is investigated as a disease, the discovery and selection of aging biomarkers and how to diagnose the aging of the organism are crucial. Here, we provide a summary of the state of the field and suggest future potential routes for research on organ senescence markers. We reviewed research on biomarkers of risk of aging from the perspective of organ aging and summarized the biomarkers currently used on three scales. We emphasize that the combination of traditional markers with emerging multifaceted biomarkers may be a better way to diagnose age-related diseases.
Collapse
Affiliation(s)
- Yulin Chen
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jiadong Li
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xinru Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| |
Collapse
|
90
|
Asanov MA, Poddubnyak AO, Sinitskaya AV, Khutornaya MV, Khryachkova OV, Sinitsky MY. Telomere Length of Cardiomyocytes in Wistar Rat Treated with Doxorubicin: In Vivo Experimental Study. Bull Exp Biol Med 2025; 178:597-600. [PMID: 40299122 DOI: 10.1007/s10517-025-06381-3] [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: 08/23/2024] [Indexed: 04/30/2025]
Abstract
The effect of doxorubicin on the relative telomere length of cardiomyocytes in Wistar rats was studied. Doxorubicin (2 mg/kg body weight) was injected into the caudal vein once a week for 4 weeks, the control group was injected with 0.9% NaCl solution in an equivalent volume. The serum concentrations of proinflammatory cytokines (MCP-1 and TNFα), oxidative stress marker 8-hydroxydeoxyguanosine (8-OHdG), and telomerase reverse transcriptase (TERT) were measured. In rats treated with doxorubicin, the relative length of cardiomyocyte telomeres and serum levels of 8-OHdG were higher than the in control. Thus, molecular effects of subchronic low-dose doxorubicin exposure in rats were revealed: telomeric regions of cardiomyocyte DNA were lengthened, and the level of oxidative stress marker increased.
Collapse
Affiliation(s)
- M A Asanov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia.
| | - A O Poddubnyak
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A V Sinitskaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - M V Khutornaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - O V Khryachkova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - M Yu Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
91
|
Gao X, Hu Y, Zhang Y, Huang Y, Zhang G, Zhang X, Zhou Y, Zhang D. A galactose-tethered tetraphenylethene prodrug mediated apoptosis of senescent cells for osteoporosis treatment. SCIENCE ADVANCES 2025; 11:eadr2833. [PMID: 39970227 PMCID: PMC11838013 DOI: 10.1126/sciadv.adr2833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Osteoporosis and bone injury healing in elderly patients are major medical challenges, often exacerbated by the accumulation of senescent cells. Herein, we show that TPE-Gal, which contains a tetraphenylethene unit and a galactose moiety, offers a promising molecular therapy designed to light up and eliminate senescent cells through a hydrolysis reaction catalyzed by β-galactosidase, an enzyme overexpressed in senescent cells. The reaction produces TPE-OH, which, in turn, increases reactive oxygen species levels within the senescent cells, leading to noninflammatory apoptosis of senescent cells. This targeted clearance mechanism helps to alleviate osteoporosis symptoms and promotes bone injury healing. Moreover, apoptotic vesicles, which are generated during the process, are partly phagocytosed by macrophages, mimicking physiological metabolic processes. This study opens new avenues for addressing bone health issues through the designed bioclearance of senescent cells, aligning with the body's natural pathways for maintaining homeostasis.
Collapse
Affiliation(s)
- Xin Gao
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yichen Hu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratories of Organic Solids and Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yingfei Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratories of Organic Solids and Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Guanxin Zhang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratories of Organic Solids and Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Deqing Zhang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratories of Organic Solids and Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
92
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
93
|
Schmieder H, Leischner C, Piotrowsky A, Marongiu L, Venturelli S, Burkard M. Exploring the link between fat-soluble vitamins and aging-associated immune system status: a literature review. Immun Ageing 2025; 22:8. [PMID: 39962579 PMCID: PMC11831837 DOI: 10.1186/s12979-025-00501-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/31/2025] [Indexed: 02/21/2025]
Abstract
The importance of vitamin D for a well-functioning immune system is becoming increasingly evident. Nevertheless, the other fat-soluble vitamins A, E and K also seem to play a central role regarding the adequate function of immune cells and to counteract excessive immune reactions and inflammatory processes. However, recognizing hidden hunger, particularly micronutrient deficiencies in vulnerable groups like the elderly, is crucial because older adults often lack sufficient micronutrients for various reasons. This review summarizes the latest findings on the immune modulating functions of fat-soluble vitamins in a physiological and pathophysiological context, provides a graphical comparison of the Recommended Daily Allowances between Deutschland, Austria, Confoederatio Helvetica (D-A-CH; eng. GSA, Germany, Switzerland, Austria), Deutsche Gesellschaft für Ernährung (DGE; eng. German Nutrition Society) and National Institutes of Health (NIH) across all age groups and, in particular, addresses the question regarding the benefits of supplementation of the respective micronutrients for the aging population of industrialized nations to strengthen the immune system. The following review highlights the importance of fat-soluble vitamins A, D, E and K which play critical roles in maintaining immune system function and, in some cases, in preventing excessive immune activation. Therefore, a better understanding of the relevance of adequate blood levels and consequently potential supplementation strategies may contribute to the prevention and management of infectious diseases as well as better overall health of the elderly.
Collapse
Affiliation(s)
- Hendrik Schmieder
- Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, Stuttgart, 70599, Germany
| | - Christian Leischner
- Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, Stuttgart, 70599, Germany
| | - Alban Piotrowsky
- Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, Stuttgart, 70599, Germany
| | - Luigi Marongiu
- Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, Stuttgart, 70599, Germany
| | - Sascha Venturelli
- Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, Stuttgart, 70599, Germany.
- Department of Vegetative and Clinical Physiology, Institute of Physiology, University of Tuebingen, Wilhelmstraße 56, Tuebingen, 72074, Germany.
| | - Markus Burkard
- Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, Stuttgart, 70599, Germany.
| |
Collapse
|
94
|
Qin Y, Liu H, Wu H. Cellular Senescence in Health, Disease, and Lens Aging. Pharmaceuticals (Basel) 2025; 18:244. [PMID: 40006057 PMCID: PMC11859104 DOI: 10.3390/ph18020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/04/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Cellular senescence is a state of irreversible cell cycle arrest that serves as a critical regulator of tissue homeostasis, aging, and disease. While transient senescence contributes to development, wound healing, and tumor suppression, chronic senescence drives inflammation, tissue dysfunction, and age-related pathologies, including cataracts. Lens epithelial cells (LECs), essential for maintaining lens transparency, are particularly vulnerable to oxidative stress-induced senescence, which accelerates lens aging and cataract formation. This review examines the dual role of senescence in LEC function and its implications for age-related cataractogenesis, alongside emerging senotherapeutic interventions. Methods: This review synthesizes findings on the molecular mechanisms of senescence, focusing on oxidative stress, mitochondrial dysfunction, and the senescence-associated secretory phenotype (SASP). It explores evidence linking LEC senescence to cataract formation, highlighting key studies on stress responses, DNA damage, and antioxidant defense. Recent advances in senotherapeutics, including senolytics and senomorphics, are analyzed for their potential to mitigate LEC senescence and delay cataract progression. Conclusions: LEC senescence is driven by oxidative damage, mitochondrial dysfunction, and impaired redox homeostasis. These factors activate senescence path-ways, including p53/p21 and p16/Rb, resulting in cell cycle arrest and SASP-mediated inflammation. The accumulation of senescent LECs reduces regenerative capacity, disrupts lens homeostasis, and contributes to cataractogenesis. Emerging senotherapeutics, such as dasatinib, quercetin, and metformin, show promise in reducing the senescent cell burden and modulating the SASP to preserve lens transparency.
Collapse
Affiliation(s)
- Ying Qin
- Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (Y.Q.); (H.L.)
| | - Haoxin Liu
- Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (Y.Q.); (H.L.)
| | - Hongli Wu
- Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (Y.Q.); (H.L.)
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
95
|
Russo T, Plessis-Belair J, Sher R, Riessland M. Regulatory Network Inference of Induced Senescent Midbrain Cell Types Reveals Cell Type-Specific Senescence-Associated Transcriptional Regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636893. [PMID: 39975267 PMCID: PMC11839108 DOI: 10.1101/2025.02.06.636893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Cellular senescence of brain cell types has become an increasingly important perspective for both aging and neurodegeneration, specifically in the context of Parkinson's Disease (PD). The characterization of classical hallmarks of senescence is a widely debated topic, whereby the context in which a senescence phenotype is being investigated, such as the cell type, the inducing stressor, and/or the model system, is an extremely important aspect to consider when defining a senescent cell. Here, we describe a cell type-specific profile of senescence through the investigation of various canonical senescence markers in five human midbrain cell lines using chronic 5-Bromodeoxyuridine (BrdU) treatment as a model of DNA damage-induced senescence. We used principal component analysis (PCA) and subsequent regulatory network inference to define both unique and common senescence profiles in the cell types investigated, as well as revealed senescence-associated transcriptional regulators (SATRs). Functional characterization of one of the identified regulators, transcription factor AP4 (TFAP4), further highlights the cell type-specificity of the expression of the various senescence hallmarks. Our data indicates that SATRs modulate cell type-specific profiles of induced senescence in key midbrain cell types that play an important role in the context of aging and PD.
Collapse
Affiliation(s)
- Taylor Russo
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Jonathan Plessis-Belair
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Roger Sher
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Markus Riessland
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
96
|
Fu TE, Zhou Z. Senescent cells as a target for anti-aging interventions: From senolytics to immune therapies. J Transl Int Med 2025; 13:33-47. [PMID: 40115034 PMCID: PMC11921816 DOI: 10.1515/jtim-2025-0005] [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] [Indexed: 03/22/2025] Open
Abstract
Aging and age-related diseases are major drivers of multimorbidity and mortality worldwide. Cellular senescence is a hallmark of aging. The accumulation of senescent cells is causally associated with pathogenesis of various age-associated disorders. Due to their promise for alleviating age-related disorders and extending healthspan, therapeutic strategies targeting senescent cells (senotherapies) as a means to combat aging have received much attention over the past decade. Among the conventionally used approaches, one is the usage of small-molecule compounds to specifically exhibit cytotoxicity toward senescent cells or inhibit deleterious effects of the senescence-associated secretory phenotype (SASP). Alternatively, there are immunotherapies directed at surface antigens specifically upregulated in senescent cells (seno-antigens), including chimeric antigen receptor (CAR) therapies and senolytic vaccines. This review gives an update of the current status in the discovery and development of senolytic therapies, and their translational progress from preclinical to clinical trials. We highlight the current challenges faced by senotherapeutic development in the context of senescence heterogeneity, with the aim of offering novel perspectives for future anti-aging interventions aimed at enhancing healthy longevity.
Collapse
Affiliation(s)
- Tianlu Esther Fu
- Faculty of Science, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| |
Collapse
|
97
|
Li S, Wang K, Wu J, Zhu Y. The immunosenescence clock: A new method for evaluating biological age and predicting mortality risk. Ageing Res Rev 2025; 104:102653. [PMID: 39746402 DOI: 10.1016/j.arr.2024.102653] [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: 09/25/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Precisely assessing an individual's immune age is critical for developing targeted aging interventions. Although traditional methods for evaluating biological age, such as the use of cellular senescence markers and physiological indicators, have been widely applied, these methods inherently struggle to capture the full complexity of biological aging. We propose the concept of an 'immunosenescence clock' that evaluates immune system changes on the basis of changes in immune cell abundance and omics data (including transcriptome and proteome data), providing a complementary indicator for understanding age-related physiological transformations. Rather than claiming to definitively measure biological age, this approach can be divided into a biological age prediction clock and a mortality prediction clock. The main function of the biological age prediction clock is to reflect the physiological state through the transcriptome data of peripheral blood mononuclear cells (PBMCs), whereas the mortality prediction clock emphasizes the ability to identify people at high risk of mortality and disease. We hereby present nearly all of the immunosenescence clocks developed to date, as well as their functional differences. Critically, we explicitly acknowledge that no single diagnostic test can exhaustively capture the intricate changes associated with biological aging. Furthermore, as these biological functions are based on the acceleration or delay of immunosenescence, we also summarize the factors that accelerate immunosenescence and the methods for delaying it. A deep understanding of the regulatory mechanisms of immunosenescence can help establish more accurate immune-age models, providing support for personalized longevity interventions and improving quality of life in old age.
Collapse
Affiliation(s)
- Shuyu Li
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ke Wang
- Department of Breast Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingni Wu
- Department of International Healthcare Center and General Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yongliang Zhu
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
98
|
Carver CM, Rodriguez SL, Atkinson EJ, Dosch AJ, Asmussen NC, Gomez PT, Leitschuh EA, Espindola-Netto JM, Jeganathan KB, Whaley MG, Kamenecka TM, Baker DJ, Haak AJ, LeBrasseur NK, Schafer MJ. IL-23R is a senescence-linked circulating and tissue biomarker of aging. NATURE AGING 2025; 5:291-305. [PMID: 39658621 PMCID: PMC11839461 DOI: 10.1038/s43587-024-00752-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/17/2024] [Indexed: 12/12/2024]
Abstract
Cellular senescence is an aging mechanism characterized by cell cycle arrest and a senescence-associated secretory phenotype (SASP). Preclinical studies demonstrate that senolytic drugs, which target survival pathways in senescent cells, can counteract age-associated conditions that span several organs. The comparative efficacy of distinct senolytic drugs for modifying aging and senescence biomarkers in vivo has not been demonstrated. Here, we established aging- and senescence-related plasma proteins and tissue transcripts that changed in old versus young female and male mice. We investigated responsivity to acute treatment with venetoclax, navitoclax, fisetin or luteolin versus transgenic senescent cell clearance in aged p16-InkAttac mice. We discovered that age-dependent changes in plasma proteins, including IL-23R, CCL5 and CA13, were reversed by senotherapeutics, which corresponded to expression differences in tissues, particularly in the kidney. In plasma from humans across the lifespan, IL-23R increased with age. Our results reveal circulating factors as candidate mediators of senescence-associated interorgan signal transduction and translationally impactful biomarkers of systemic senescent cell burden.
Collapse
Affiliation(s)
- Chase M Carver
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Sonia L Rodriguez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth J Atkinson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Dosch
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Niels C Asmussen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Paul T Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Ethan A Leitschuh
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Jair M Espindola-Netto
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Madison G Whaley
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, UF Scripps Institute, The Scripps Research Institute, Scripps Florida, Jupiter, FL, USA
| | - Darren J Baker
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Nathan K LeBrasseur
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
- Department of Neuroscience, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
99
|
Yasuda T, Alan Wang Y. Immune therapeutic strategies for the senescent tumor microenvironment. Br J Cancer 2025; 132:237-244. [PMID: 39468331 PMCID: PMC11790855 DOI: 10.1038/s41416-024-02865-7] [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: 06/26/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024] Open
Abstract
Senescent cells can either to promote immunosuppressive tumor microenvironment or facilitate immune surveillance. Despite the revolutionary impact of cancer immunotherapy, durable responses in solid tumors, particularly in advanced stages, remain limited. Recent studies have shed light on the influence of senescent status within the tumor microenvironment (TME) on therapy resistance and major efforts are needed to overcome these challenges. This review summarizes recent advancements in targeting cellular senescence, with a particular focus on immunomodulatory approaches on the hallmarks of cellular senescence.
Collapse
Affiliation(s)
- Tadahito Yasuda
- Brown Center for Immunotherapy, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA.
| | - Y Alan Wang
- Brown Center for Immunotherapy, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center Indianapolis, Indianapolis, USA
| |
Collapse
|
100
|
Costa CM, Pedrosa SS, Kirkland JL, Reis F, Madureira AR. The senotherapeutic potential of phytochemicals for age-related intestinal disease. Ageing Res Rev 2025; 104:102619. [PMID: 39638096 DOI: 10.1016/j.arr.2024.102619] [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: 09/17/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
During the last few decades, life expectancy has increased worldwide along with the prevalence of several age-related diseases. Among aging pathways, cellular senescence and chronic inflammation (or "inflammaging") appear to be connected to gut homeostasis and dysbiosis of the microbiome. Cellular senescence is a state of essentially irreversible cell cycle arrest that occurs in response to stress. Although senescent cells (SC) remain metabolically active, they do not proliferate and can secrete inflammatory and other factors comprising the senescence-associated secretory phenotype (SASP). Accumulation of SCs has been linked to onset of several age-related diseases, in the brain, bones, the gastrointestinal tract, and other organs and tissues. The gut microbiome undergoes substantial changes with aging and is tightly interconnected with either successful (healthy) aging or disease. Senotherapeutic drugs are compounds that can clear senescent cells or modulate the release of SASP factors and hence attenuate the impact of the senescence-associated pro-inflammatory state. Phytochemicals, phenolic compounds and terpenes, which have antioxidant and anti-inflammatory activities, could also be senotherapeutic given their ability to act upon senescence-linked cellular pathways. The aim of this review is to dissect links among the gut microbiome, cellular senescence, inflammaging, and disease, as well as to explore phytochemicals as potential senotherapeutics, focusing on their interactions with gut microbiota. Coordinated targeting of these inter-related processes might unveil new strategies for promoting healthy aging.
Collapse
Affiliation(s)
- Célia Maria Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| | - Sílvia Santos Pedrosa
- Biorbis, Unipessoal LDA, Edifício de Biotecnologia da Universidade Católica Portuguesa, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| | - James L Kirkland
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA.
| | - Flávio Reis
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3000-548, Portugal; Clinical Academic Center of Coimbra, Coimbra 3004-531, Portugal.
| | - Ana Raquel Madureira
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| |
Collapse
|