1
|
Wang T, Shi X, Xu X, Zhang J, Ma Z, Meng C, Jiao D, Wang Y, Chen Y, He Z, Zhu Y, Liu HN, Zhang T, Jiang Q. Emerging prodrug and nano-drug delivery strategies for the detection and elimination of senescent tumor cells. Biomaterials 2025; 318:123129. [PMID: 39922127 DOI: 10.1016/j.biomaterials.2025.123129] [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: 11/04/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/10/2025]
Abstract
Tumor cellular senescence, characterized by reversible cell cycle arrest following anti-cancer therapies, presents a complex paradigm in oncology. Given that senescent tumor cells may promote angiogenesis, tumorigenesis, and metastasis, selective killing senescent cells (SCs)-a strategy termed senotherapy-has emerged as a promising approach to improve cancer treatment. However, the clinical implementation of senotherapy faces significant hurdles, including lack of precise methods for SCs identification and the potential for adverse effects associated with highly cytotoxic senolytic agents. In this account, we elucidate recent advancement in developing novel approaches for the detection and selective elimination of SCs, encompassing prodrugs, nanoparticles, and other cutting-edge drug delivery systems such as PROTAC technology and CAR T cell therapy. Furthermore, we explore the paradoxical nature of SCs, which can induce growth arrest in adjacent neoplastic cells and recruit immunomodulatory cells that contribute to tumor suppression. Therefore, we utilize SCs membrane as vehicles to elicit antitumor immunity and potentially augment existing anti-cancer therapies. Finally, the opportunities and challenges are put forward to facilitate the development and clinical transformation of SCs detection, elimination or utilization.
Collapse
Affiliation(s)
- Tao Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Xiaolan Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jiaming Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhengdi Ma
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chen Meng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dian Jiao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yubo Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yanfei Chen
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, 110002, China.
| | - He-Nan Liu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Tianhong Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Qikun Jiang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China; Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Medical University, Haikou, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
2
|
Xie J, Shu X, Xie Z, Tang J, Wang G. Pharmacological modulation of cellular senescence: Implications for breast cancer progression and therapeutic strategies. Eur J Pharmacol 2025; 997:177475. [PMID: 40049574 DOI: 10.1016/j.ejphar.2025.177475] [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: 12/09/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Abstract
Senescence, defined by the cessation of cell proliferation, plays a critical and multifaceted role in breast cancer progression and treatment. Senescent cells produce senescence-associated secretory phenotypes (SASP) comprising inflammatory cytokines, chemokines, and small molecules, which actively shape the tumor microenvironment, influencing cancer development, progression, and metastasis. This review provides a comprehensive analysis of the types and origins of senescent cells in breast cancer, alongside their markers and detection methods. Special focus is placed on pharmacological strategies targeting senescence, including drugs that induce or inhibit senescence, their molecular mechanisms, and their roles in therapeutic outcomes when combined with chemotherapy and radiotherapy. By exploring these pharmacological interventions and their impact on breast cancer treatment, this review underscores the potential of senescence-targeting therapies to revolutionize breast cancer management.
Collapse
Affiliation(s)
- Jialing Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Xianlong Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Zilan Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| |
Collapse
|
3
|
Cong Y, Li X, Hong H. Current strategies for senescence treatment: Focused on theranostic performance of nanomaterials. J Control Release 2025; 382:113710. [PMID: 40220869 DOI: 10.1016/j.jconrel.2025.113710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
Age-related diseases imposed heavy burdens to the healthcare systems globally, while cell senescence served as one fundamental molecular/cellular basis for these diseases. How to tackle the senescence-relevant problems is a hotspot for biomedical research. In this review article, the hallmarks and molecular pathways of cell senescence were firstly discussed, followed by the introduction of the current anti-senescence strategies, including senolytics and senomorphics. With suitable physical or chemical properties, multiple types of nanomaterials were used successfully in senescence therapeutics, as well as senescence detection. Based on the accumulating knowledges for senescence, the rules of how to use these nanoplatforms more efficiently against senescence were also summarized, including but not limited to surface modification, material-cargo interactions, factor responsiveness etc. The comparison of these "senescence-selective" nanoplatforms to other treatment options (prodrugs, ADCs, PROTACs, CART etc.) was also given. Learning from the past, nanotechnology can add more choice for treating age-related diseases, and provide more (diagnostic) information to further our understanding of senescence process.
Collapse
Affiliation(s)
- Yiyang Cong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China
| | - Xiaoyang Li
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China.
| |
Collapse
|
4
|
Ahmadizad Firouzjaei A, Aghaee-Bakhtiari SH. Integrating cuproptosis and immunosenescence: A novel therapeutic strategy in cancer treatment. Biochem Biophys Rep 2025; 42:101983. [PMID: 40224540 PMCID: PMC11986980 DOI: 10.1016/j.bbrep.2025.101983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/01/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Recent advancements in our understanding of cell death mechanisms have progressed beyond traditional apoptosis to encompass various forms of regulated cell death, notably cuproptosis. This copper-dependent cell death occurs when copper interacts with lipoylated enzymes in the tricarboxylic acid cycle, leading to protein aggregation and subsequent cell death. Alongside this, immunosenescence the gradual decline in immune function due to aging has emerged as a significant factor in cancer progression and response to treatment. Innovative strategies that integrate cuproptosis and immunosenescence are showing considerable promise in cancer therapy. By leveraging the altered copper metabolism in cancer cells, cuproptosis can selectively induce cell death, effectively targeting and eliminating tumors. Simultaneously, addressing immunosenescence can rejuvenate the aging immune system, enhancing its capacity to identify and destroy cancer cells. This dual approach creates a synergistic effect, optimizing therapeutic efficacy by directly attacking tumor cells while revitalizing the immune response. Such integration bolsters the defense against cancer progression and recurrence and holds great potential for advancing cancer treatment modalities and improving patient outcomes. This paper delves into the interactions between cuproptosis and immunosenescence, emphasizing their implications for developing innovative cancer therapies.
Collapse
Affiliation(s)
- Ali Ahmadizad Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Aswani BS, Sajeev A, Hegde M, Mishra A, Abbas M, Vayalpurayil T, Sethi G, Kunnumakkara AB. Exosomal dynamics: Bridging the gap between cellular senescence and cancer therapy. Mech Ageing Dev 2025; 225:112045. [PMID: 40074065 DOI: 10.1016/j.mad.2025.112045] [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: 11/28/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Cancer remains one of the most devastating diseases, severely affecting public health and contributing to economic instability. Researchers worldwide are dedicated to developing effective therapeutics to target cancer cells. One promising strategy involves inducing cellular senescence, a complex state in which cells exit the cell cycle. Senescence has profound effects on both physiological and pathological processes, influencing cellular systems through secreted factors that affect surrounding and distant cells. Among these factors are exosomes, small extracellular vesicles that play crucial roles in cellular communication, development, and defense, and can contribute to pathological conditions. Recently, there has been increasing interest in engineering exosomes as precise drug delivery vehicles, capable of targeting specific cells or intracellular components. Studies have emphasized the significant role of exosomes from senescent cells in cancer progression and therapy. Notably, chemotherapeutic agents can alter the tumor microenvironment, induce senescence, and trigger immune responses through exosome-mediated cargo transfer. This review explores the intricate relationship between cellular senescence, exosomes, and cancer, examining how different therapeutics can eliminate cancer cells or promote drug resistance. It also investigates the molecular mechanisms and signaling pathways driving these processes, highlighting current challenges and proposing future perspectives to uncover new therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Babu Santha Aswani
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Anjana Sajeev
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Anamika Mishra
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Thafasalijyas Vayalpurayil
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India.
| |
Collapse
|
6
|
Fang H, Yu E, Liu C, Eapen C, Cheng C, Hu T. Metabolic landscape and rewiring in normal hematopoiesis, leukemia and aging. Semin Cancer Biol 2025; 111:1-15. [PMID: 39933639 DOI: 10.1016/j.semcancer.2025.02.003] [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: 10/23/2024] [Revised: 01/06/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Recent advancements in metabolism research have demonstrated its critical roles in a lot of critical biological processes, including stemness maintenance, cell differentiation, proliferation, and function. Hematopoiesis is the fundamental cell differentiation process with the production of millions of red blood cells per second in carrying oxygen and white blood cells in fighting infection and cancers. The differentiation processes of hematopoietic stem and progenitor cells (HSPCs) are accompanied by significant metabolic reprogramming. In hematological malignancy, metabolic reprogramming is also essential to the malignant hematopoiesis processes. The metabolic rewiring is driven by distinct molecular mechanisms that meet the specific demands of different target cells. Leukemic cells, for instance, adopt unique metabolic profiles to support their heightened energy needs for survival and proliferation. Moreover, aging HSPCs exhibit altered energy consumption compared to their younger counterparts, often triggering protective mechanisms at the cellular level. In this review, we provide a comprehensive analysis of the metabolic processes involved in hematopoiesis and the metabolic rewiring that occurs under adverse conditions. In addition, we highlight current research directions and discuss the potential of targeting metabolic pathways for the management of hematological malignancies and aging.
Collapse
Affiliation(s)
- Hui Fang
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Enze Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa 999078, Macao
| | - Chang Liu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Christy Eapen
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States
| | - Chunming Cheng
- Stephenson Cancer Center at Oklahoma University, Oklahoma City, OK 73104, United States.
| | - Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, United States.
| |
Collapse
|
7
|
Abe H, Indo HP, Ito H, Majima HJ, Tanaka T. Synephrine Inhibits Oxidative Stress and H 2O 2-Induced Premature Senescence. Cell Biochem Biophys 2025; 83:2607-2622. [PMID: 39832117 PMCID: PMC12089197 DOI: 10.1007/s12013-025-01669-7] [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] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Synephrine, a protoalkaloid found in Citrus aurantium (CA) peels, exerts lipolytic, anti-inflammatory, and vasoconstrictive effects; however, its antioxidant activity remains unclear. In this study, electron spin resonance spectroscopy revealed that synephrine scavenged both hydroxyl and superoxide anion radicals. Several external stimuli, such as H2O2, X-rays, and ultraviolet (UV) radiation, cause stress-induced premature senescence (SIPS). As oxidative stress induces SIPS, we hypothesized that synephrine, an antioxidant, would suppress H2O2-induced premature senescence in WI-38 cells. Synephrine significantly decreased the reactive oxygen species levels induced by H2O2, thereby reducing lipid peroxidation, and oxidative DNA damage and preventing SIPS. Additionally, synephrine inhibited mitochondrial dysfunction in H2O2-treated WI-38 cells. The expression levels of p53, p21, and p16-INK4A, which are involved in the induction of cell cycle arrest in SIPS, were significantly lower in synephrine-treated cells than in untreated cells. Our results indicate that synephrine inhibits H2O2-induced oxidative stress and mitochondrial dysfunction, suppressing premature senescence by inhibiting activation of the p53-p21 and p16-INK4A-pRB pathways.
Collapse
Affiliation(s)
- Hiroshi Abe
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
| | - Hiroko P Indo
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan.
| | - Hiromu Ito
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
- Quantum RedOx Chemistry Team, Quantum Life Spin Group, Institute for Quantum Life Science (iQLS), National Institutes for Quantum Science and Technology (QST), Chiba, 263-8555, Japan
| | - Hideyuki J Majima
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Tatsuro Tanaka
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
| |
Collapse
|
8
|
Picos A, Seoane N, Campos-Toimil M, Viña D. Vascular senescence and aging: mechanisms, clinical implications, and therapeutic prospects. Biogerontology 2025; 26:118. [PMID: 40418230 DOI: 10.1007/s10522-025-10256-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
The aging vasculature is characterized by endothelial dysfunction, arterial stiffness, and increased susceptibility to vascular pathologies. Central to these changes is the process of cellular senescence, where endothelial and vascular smooth muscle cells lose their replicative and functional capacity and adopt a pro-inflammatory secretory phenotype. This review provides an overview of the key mechanisms underlying vascular senescence, including the p53/p21 and p16/Rb pathways, the senescence-associated secretory phenotype (SASP), and oxidative stress, examines its contribution to cardiovascular diseases in older adults, and highlights emerging therapeutic strategies aimed at delaying or reversing these age-related vascular changes. In vascular cells, DNA damage, oxidative stress, and chronic inflammation associated with aging converge to amplify senescence. Clinically, vascular senescence is linked with hypertension, atherosclerosis, and increased overall cardiovascular risk. Several interventions, ranging from senolytics to lifestyle factors, show promise in mitigating these changes; however, long-term studies are needed. Given that vascular senescence is a pivotal driver of cardiovascular pathology in aging, targeting senescent cells or their secretory phenotype may potentially offer new avenues for preventing or attenuating age-related vascular diseases. This review presents an updated and integrative overview of vascular senescence, connecting fundamental cellular mechanisms with their clinical manifestations and highlighting the most promising therapeutic interventions.
Collapse
Affiliation(s)
- Aitor Picos
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| | - Nuria Seoane
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Manuel Campos-Toimil
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| | - Dolores Viña
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Translational Research in Neurological Diseases (ITEN), Health Research Institute of Santiago de Compostela (IDIS), USC University Hospital Complex (CHUS), SERGAS, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| |
Collapse
|
9
|
Li XR, Qi L, Zhang XW, Wei C, Yu B, Pei TL. Quercetin and Nano-Derivatives: Potential and Challenges in Cancer Therapy. Int J Nanomedicine 2025; 20:6701-6720. [PMID: 40444010 PMCID: PMC12120254 DOI: 10.2147/ijn.s509877] [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: 12/02/2024] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Quercetin, a prevalent flavonol compound, has gained attention for its multifaceted mechanisms of action against various cancers, highlighting its potential as an adjunctive therapy in cancer treatments. This review aims to systematically evaluate the structural optimization, mechanisms of action, and clinical applications of quercetin and its nano-derivatives in cancer treatment. Employing a bibliometric analysis of 6231 articles from the Web of Science Core Collection, we observed a notable increase in annual publications, particularly from the USA and China, indicating a growing interest in quercetin's therapeutic potential. Our findings reveal that quercetin enhances the efficacy of conventional therapies by modulating critical signaling pathways, thereby increasing cancer cell sensitivity while simultaneously protecting normal tissues from therapy-induced damage. Structural modifications, including glycosylation, methylation, sulfation, and glucuronidation, alongside nanoparticle formulation, significantly improve the stability, solubility, and bioavailability of quercetin, enabling targeted drug delivery. Despite the promising preclinical outcomes, the clinical translation of quercetin remains nascent, necessitating further rigorous research to validate its safety and efficacy in human subjects. In conclusion, while quercetin exhibits substantial anticancer properties and therapeutic potential, future studies should focus on expanding sample sizes, elucidating metabolic pathways, and conducting comprehensive clinical trials to inform its application in oncology.
Collapse
Affiliation(s)
- Xin-Ru Li
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, Shandong Province, 272000, People’s Republic of China
| | - Lin Qi
- Affiliated Hospital of Jining Medical University, Jining, 272000, People’s Republic of China
| | - Xi-Wen Zhang
- College of The Second Clinical Medical, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, 450003, People’s Republic of China
| | - Chao Wei
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, Shandong Province, 272000, People’s Republic of China
| | - Bin Yu
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, Shandong Province, 272000, People’s Republic of China
| | - Tian-Li Pei
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, 550000, People’s Republic of China
| |
Collapse
|
10
|
Stojanović SD, Thum T, Bauersachs J. Anti-senescence therapies: a new concept to address cardiovascular disease. Cardiovasc Res 2025; 121:730-747. [PMID: 40036821 PMCID: PMC12101330 DOI: 10.1093/cvr/cvaf030] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/16/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Accumulation of senescent cells is an increasingly recognized factor in the development and progression of cardiovascular (CV) disease (CVD). Senescent cells of different types display a pro-inflammatory and matrix remodelling molecular programme, known as the 'senescence-associated secretory phenotype' (SASP), which has roots in (epi)genetic changes. Multiple therapeutic options (senolytics, anti-SASP senomorphics, and epigenetic reprogramming) that delete or ameliorate cellular senescence have recently emerged. Some drugs routinely used in the clinics also have anti-senescence effects. However, multiple challenges hinder the application of novel anti-senescence therapeutics in the clinical setting. Understanding the biology of cellular senescence, advantages and pitfalls of anti-senescence treatments, and patients who can profit from these interventions is necessary to introduce this novel therapeutic modality into the clinics. We provide a guide through the molecular machinery of senescent cells, systematize anti-senescence treatments, and propose a pathway towards senescence-adapted clinical trial design to aid future efforts.
Collapse
Affiliation(s)
- Stevan D Stojanović
- Department of Cardiology and Angiology, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- PRACTIS Clinician Scientist Program, Dean’s Office for Academic Career Development, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| |
Collapse
|
11
|
Wu J, Zhang L, Zhao Z, Liu Y, Li Z, Feng X, Zhang L, Yao X, Du J, Chen L, Zhou Z. Advancing T-cell immunotherapy for cellular senescence and disease: Mechanisms, challenges, and clinical prospects. Ageing Res Rev 2025; 109:102783. [PMID: 40412763 DOI: 10.1016/j.arr.2025.102783] [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: 03/26/2025] [Revised: 05/12/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Cellular senescence is a complex biological process with a dual role in tissue homeostasis and aging-related pathologies. Accumulation of senescent cells promotes chronic inflammation, tissue dysfunction, age-related diseases, and tumor suppression. Recent advancements in immunotherapy have positioned T cell-based approaches as precision tools for the targeted clearance of senescent cells, offering a novel avenue for anti-aging interventions. This review explores the molecular mechanisms underlying cellular senescence, focusing on its immunogenic features and interactions with T cells, including T-cell activation, antigen recognition, modulation of tumor microenvironment (TME), and immune evasion strategies. Innovations such as chimeric antigen receptor (CAR)-T cells, immune checkpoint therapies, and SASP-neutralizing approaches are highlighted as breakthrough strategies for enhancing senescent cell eradication. The integration of multi-omics and artificial intelligence is further catalyzing the development of personalized therapies to amplify immune surveillance and tissue rejuvenation. Clinically, T cell-based interventions hold promise for mitigating age-related pathologies and extending healthspan, yet challenges remain in optimizing target specificity, countering immunosuppressive niches, and overcoming immune senescence in aging populations. This review synthesizes current advances and challenges, highlighting the potential of T cell immunotherapy as a cornerstone of anti-aging medicine and emphasizing the need for interdisciplinary innovation to translate preclinical findings into transformative therapies for aging and age-related diseases.
Collapse
Affiliation(s)
- Jizhun Wu
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lu Zhang
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Zihan Zhao
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yuping Liu
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhengxing Li
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiaohang Feng
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lin Zhang
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiang Yao
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China.
| | - Zhuolong Zhou
- Department of Colorectal Surgery, The Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Biomedical Sciences, College of Medicine and Veterinary Medicine, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
12
|
Ryan P, Lee J. In vitro senescence and senolytic functional assays. Biomater Sci 2025. [PMID: 40375674 DOI: 10.1039/d4bm01684j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
A detailed understanding of aging biology and the development of anti-aging therapeutic strategies remain imperative yet inherently challenging due to the protracted nature of aging. Cellular senescence arises naturally through replicative exhaustion and is accelerated by clinical treatments or environmental stressors. The accumulation of senescent cells-defined by a loss of mitogenic potential, resistance to apoptosis, and acquisition of a pro-inflammatory secretory phenotype-has been implicated as a key driver of chronic disease, tissue degeneration, and organismal aging. Recent studies have highlighted the therapeutic promise of senolytic drugs, which selectively eliminate senescent cells. Compelling results from preclinical animal studies and ongoing clinical trials underscore this potential. However, the clinical translation of senolytics requires further pharmacological validation to refine selectivity, minimize toxicity, and determine optimal dosing. Equally important is the evaluation of senolytics' potential to restore tissue structure and function by reducing the senescent cell burden. In vitro tissue culture models offer a powerful platform to advance these efforts. This review summarizes the current landscape of in vitro systems used for inducing cellular senescence-referred to as "senescence assays"-and for screening senolytic drugs-referred to as "senolytic assays". We conclude by discussing key challenges to improving mechanistic insight, predictive accuracy, and clinical relevance in senolytic drug development, as well as emerging applications of senolytic therapies.
Collapse
Affiliation(s)
- Patrick Ryan
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, 01003, USA.
| | - Jungwoo Lee
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, 01003, USA.
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
13
|
Saleh T, Greenberg EF, Faber AC, Harada H, Gewirtz DA. A Critical Appraisal of the Utility of Targeting Therapy-Induced Senescence for Cancer Treatment. Cancer Res 2025; 85:1755-1768. [PMID: 40036150 DOI: 10.1158/0008-5472.can-24-2219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/03/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025]
Abstract
Cancer chemotherapy and radiotherapy are rarely successful in eliminating the entire tumor population, often leaving behind a subpopulation of senescent cells that can contribute to disease recurrence. These senescent tumor cells also secrete various chemokines and cytokines that may be tumor promoting and immunosuppressive. Recognition of the deleterious impact of therapy-induced senescence has led to the preclinical development of senolytic compounds that eliminate senescent cells, representing a potential strategy to enhance the efficacy of conventional and targeted anticancer therapy. However, it remains uncertain whether this strategy can or will be translated to the clinic. This review provides a summary of the recent preclinical literature supporting the use of senolytics as an adjunct for cancer treatment, discusses the limitations associated with their use in the current preclinical models, and provides perspectives on the clinical development of senolytics in cancer treatment regimens. Overall, preclinical studies support the potential of senolytics to enhance efficacy and prolong the antitumor activity of current standard-of-care cancer therapies that promote senescence. However, further work is needed to develop optimal senolytic agents with the appropriate combination of properties for clinical testing, specifically, activity in the context of therapy-induced senescence with acceptable tolerability.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | | | - Anthony C Faber
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia
- Department of Pediatrics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Hisashi Harada
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - David A Gewirtz
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
14
|
Chen C, Wang L. Aging and metabolic dysfunction-associated steatotic liver disease: a bidirectional relationship. Front Med 2025:10.1007/s11684-025-1133-7. [PMID: 40316793 DOI: 10.1007/s11684-025-1133-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/09/2025] [Indexed: 05/04/2025]
Abstract
In recent years, aging and cellular senescence have triggered an increased interest in corresponding research fields. Evidence shows that the complex aging process is involved in the development of many chronic liver diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). In fact, aging has a tremendous effect on the liver, leading to a gradual decline in the metabolism, detoxification and immune functions of the liver, which in turn increases the risk of liver disease. These changes can be based on the aging of liver cells (hepatocytes, liver sinusoidal endothelial cells, hepatic stellate cells, and Kupffer cells). Similarly, patients with liver diseases exhibit increases in the aging phenotype and aging cells, often manifesting as faster physical functional decline, which is closely related to the promoting effect of liver disease on aging. This review summarizes the interplay between MASLD/MASH development and aging, aiming to reveal the complex relationships that exacerbate one another. Moreover, the corresponding schemes for delaying aging or treating diseases are discussed to provide a basis for the development of effective prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
15
|
Xiong J, Guo Q, Luo X. Cellular senescence in age-related musculoskeletal diseases. Front Med 2025:10.1007/s11684-025-1125-7. [PMID: 40314896 DOI: 10.1007/s11684-025-1125-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/16/2024] [Indexed: 05/03/2025]
Abstract
Aging is typically associated with decreased musculoskeletal function, leading to reduced mobility and increased frailty. As a hallmark of aging, cellular senescence plays a crucial role in various age-related musculoskeletal diseases, including osteoporosis, osteoarthritis, intervertebral disc degeneration, and sarcopenia. The detrimental effects of senescence are primarily due to impaired regenerative capacity of stem cells and the pro-inflammatory environment created by accumulated senescent cells. The secreted senescence-associated secretory phenotype (SASP) can induce senescence in neighboring cells, further amplifying senescent signals. Although the removal of senescent cells and the suppression of SASP factors have shown promise in alleviating disease progression and restoring musculoskeletal health in mouse models, clinical trials have yet to demonstrate significant efficacy. This review summarizes the mechanisms of cellular senescence in age-related musculoskeletal diseases and discusses potential therapeutic strategies targeting cellular senescence.
Collapse
Affiliation(s)
- Jinming Xiong
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Qiaoyue Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| |
Collapse
|
16
|
Donovan LJ, Brewer CL, Bond SF, Laslavic AM, Pena Lopez A, Colman L, Jordan CE, Hansen LH, González OC, Pujari A, de Lecea L, Quarta M, Kauer JA, Tawfik VL. Aging and injury drive neuronal senescence in the dorsal root ganglia. Nat Neurosci 2025; 28:985-997. [PMID: 40369367 PMCID: PMC12081305 DOI: 10.1038/s41593-025-01954-x] [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: 01/12/2024] [Accepted: 03/21/2025] [Indexed: 05/16/2025]
Abstract
Aging negatively impacts central nervous system function; however, there is limited information about the cellular impact of aging on peripheral nervous system function. Importantly, injury to vulnerable peripheral axons of dorsal root ganglion (DRG) neurons results in somatosensory dysfunction, such as pain, at higher rates in aged individuals. Cellular senescence is common to both aging and injury and contributes to the aged pro-inflammatory environment. We discovered DRG neuron senescence in the context of aging and pain-inducing peripheral nerve injury in young (~3 months) and aged (~24 months) male and female mice. Senescent neurons were dynamic and heterogeneous in their expression of multiple senescence markers, including pro-inflammatory factor IL6. Senescence marker-expressing neurons had nociceptor-like profiles, included high-firing phenotypes and displayed increased excitability after IL6 application. Furthermore, elimination of senescent cells resulted in improvement of nociceptive behaviors in nerve-injured mice. Finally, male and female post-mortem human DRG contained senescent neurons that increased with age (~32 years old versus 65 years old). Overall, we describe a susceptibility of the peripheral nervous system to neuronal senescence-a potential targetable mechanism to treat sensory dysfunction, such as chronic pain, particularly in aged populations.
Collapse
Affiliation(s)
- Lauren J Donovan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA.
| | - Chelsie L Brewer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sabrina F Bond
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | | | - Aleishai Pena Lopez
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Laura Colman
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Claire E Jordan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Linus H Hansen
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Oscar C González
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | | | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | | | - Julie A Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
17
|
Jochems F, Baltira C, MacDonald JA, Daniels V, Mathur A, de Gooijer MC, van Tellingen O, Letai A, Bernards R. Senolysis by ABT-263 is associated with inherent apoptotic dependence of cancer cells derived from the non-senescent state. Cell Death Differ 2025; 32:855-865. [PMID: 39706991 DOI: 10.1038/s41418-024-01439-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
Cellular senescence is a stress response that cells can employ to resist cell death. Senescent cells rely on anti-apoptotic signaling for their survival, which can be targeted by senolytic agents, like the BCL-XL, BCL-2, BCL-W inhibitor ABT-263. However, the response to ABT-263 of senescent cancer cells ranges from highly sensitive to refractory. Using BH3 profiling, we identify here apoptotic blocks in cancer cells that are resistant to this senolytic treatment and discover a correlation between mitochondrial apoptotic priming and cellular sensitivity to ABT-263 in senescence. Intriguingly, ABT-263 sensitivity correlates with overall mitochondrial apoptotic priming, not only in senescence but also in the parental state. Moreover, we confirm that ABT-263 exposure increases dependency on MCL-1, which is most enhanced in ABT-263 sensitive cells. ABT-263 resistant cells however upregulate MCL-1, while sensitive cells exhibit low levels of this anti-apoptotic protein. Overall, our data indicate that the response of senescent cells to ABT-263 is predetermined by the mitochondrial apoptotic priming state of the parental cells, which could serve as a predictive biomarker for response to senolytic therapy.
Collapse
Affiliation(s)
- Fleur Jochems
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Chrysiida Baltira
- Division of Pharmacology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Julie A MacDonald
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Veerle Daniels
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Abhijeet Mathur
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
- Faculty of Biology, Medicine and Health, University of Manchester; The Christie NHS Foundation Trust, Manchester, UK
| | - Olaf van Tellingen
- Division of Pharmacology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Anthony Letai
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.
| |
Collapse
|
18
|
Rys RN, Calcinotto A. Senescent neutrophils: a hidden role in cancer progression. Trends Cell Biol 2025; 35:399-411. [PMID: 39362804 DOI: 10.1016/j.tcb.2024.09.001] [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: 06/03/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Neutrophils have recently received increased attention in cancer because they contribute to all stages of cancer. Neutrophils are so far considered to have a short half-life. However, a growing body of literature has shown that tumor-associated neutrophils (TANs) acquire a prolonged lifespan. This review discusses recent work surrounding the mechanisms by which neutrophils can persist in the tumor microenvironment (TME). It also highlights different scenarios for therapeutic targeting of protumorigenic neutrophils, supporting the idea that, in tumors, inhibition of neutrophil recruitment is not sufficient because these cells can persist and remain hidden from current interventions. Hence, the elimination of long-lived neutrophils should be pursued to increase the efficacy of standard therapy.
Collapse
Affiliation(s)
- Ryan N Rys
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Arianna Calcinotto
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland.
| |
Collapse
|
19
|
Tanaka S, Mifune Y, Inui A, Yamaura K, Furukawa T, Kato T, Kusunose M, Matsumoto T, Matsushita T, Kuroda R. Mitochondrial Dysfunction of the Subsynovial Connective Tissue in Patients With Carpal Tunnel Syndrome. J Orthop Res 2025; 43:1045-1053. [PMID: 40099548 DOI: 10.1002/jor.26064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
In idiopathic carpal tunnel syndrome (CTS), fibrosis and thickening of the subsynovial connective tissue (SSCT) increase pressure within the carpal tunnel, resulting in median nerve entrapment. Mitochondrial dysfunction in tissues reportedly leads to senescent cell accumulation and various diseases through reduced adenosine triphosphate (ATP) and excessive reactive oxygen species (ROS) production; however, no reports have linked this to CTS. Therefore, this study aimed to evaluate mitochondrial function in SSCTs of patients with CTS. This study investigated SSCTs obtained during carpal tunnel release surgery in patients with CTS (CTS group) and those obtained during tendon transfer or tendon rupture surgery in patients without CTS (control group) from April 2021 to March 2023 at our hospital. Outcome measures included superoxide dismutase (SOD) activity, gene expression levels, immunofluorescence staining, ATP production assays, and transmission electron microscopy (TEM). p values were calculated using the Mann-Whitney U test. The CTS and control groups included 10 and 5 patients (mean age, 67.8 ± 9.57 and 65.4 ± 9.75 years), respectively. The CTS group exhibited decreased SOD activity (p = 0.026), increased gene expression of mitochondrial biosynthetic factors, and decreased mitochondrial ATP production (p = 0.027). The CTS group showed increased mitochondrial ROS production (p = 0.038) on immunofluorescence and larger mitochondrial area (p = 0.030) and fewer mitochondrial cristae (p = 0.045) on TEM. Multiple mitochondrial function assays suggested mitochondrial dysfunction of SSCTs in the CTS group. STATEMENT OF CLINICAL SIGNIFICANCE: This research provided important information on the histological changes in the subsynovial connective tissue within the carpal tunnel in carpal tunnel syndrome.
Collapse
Affiliation(s)
- Shuya Tanaka
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Yutaka Mifune
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Atsuyuki Inui
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Kohei Yamaura
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Takahiro Furukawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Tatsuo Kato
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Masaya Kusunose
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kobe University, Kobe-shi, Hyogo, Japan
| |
Collapse
|
20
|
Zhang Z, Yang R, Zi Z, Liu B. A new clinical age of aging research. Trends Endocrinol Metab 2025; 36:440-458. [PMID: 39227191 DOI: 10.1016/j.tem.2024.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024]
Abstract
Aging is a major risk factor for a variety of diseases, thus, translation of aging research into practical applications is driven by the unmet need for existing clinical therapeutic options. Basic and translational research efforts are converging at a critical stage, yielding insights into how fundamental aging mechanisms are used to identify promising geroprotectors or therapeutics. This review highlights several research areas from a clinical perspective, including senescent cell targeting, alleviation of inflammaging, and optimization of metabolism with endogenous metabolites or precursors. Refining our understanding of these key areas, especially from the clinical angle, may help us to better understand and attenuate aging processes and improve overall health outcomes.
Collapse
Affiliation(s)
- Zhen Zhang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518055, China
| | - Renlei Yang
- Department of Plastic Surgery, Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Zhike Zi
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518055, China.
| |
Collapse
|
21
|
MacDonald JA, Bradshaw GA, Jochems F, Bernards R, Letai A. Apoptotic priming in senescence predicts specific senolysis by quantitative analysis of mitochondrial dependencies. Cell Death Differ 2025; 32:802-817. [PMID: 39762561 DOI: 10.1038/s41418-024-01431-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 05/21/2025] Open
Abstract
Cellular senescence contributes to a variety of pathologies associated with aging and is implicated as a cellular state in which cancer cells can survive treatment. Reported senolytic drug treatments act through varying molecular mechanisms, but heterogeneous efficacy across the diverse contexts of cellular senescence indicates a need for predictive biomarkers of senolytic activity. Using multi-parametric analyses of commonly reported molecular features of the senescent phenotype, we assayed a variety of models, including malignant and nonmalignant cells, using several triggers of senescence induction and found little univariate predictive power of these traditional senescence markers to identify senolytic drug sensitivity. We sought to identify novel drug targets in senescent cells that were insensitive to frequently implemented senolytic therapies, such as Navitoclax (ABT-263), using quantitative mass spectrometry to measure changes in the senescent proteome, compared to cells which acquire an acute sensitivity to ABT-263 with senescence induction. Inhibition of the antioxidant GPX4 or the Bcl-2 family member MCL-1 using small molecule compounds in combination with ABT-263 significantly increased the induction of apoptosis in some, but not all, previously insensitive senescent cells. We then asked if we could use BH3 profiling to measure differences in mitochondrial apoptotic priming in these models of cellular senescence and predict sensitivity to the senolytics ABT-263 or the combination of dasatinib and quercetin (D + Q). We found, despite being significantly less primed for apoptosis overall, the dependence of senescent mitochondria on BCL-XL was significantly correlated to senescent cell killing by both ABT-263 and D + Q, despite no significant changes in the gene or protein expression of BCL-XL. However, our data caution against broad classification of drugs as globally senolytic and instead provide impetus for context-specific senolytic targets and propose BH3 profiling as an effective predictive biomarker.
Collapse
Affiliation(s)
- Julie A MacDonald
- Dana Farber Cancer Institute, Boston, MA, USA
- Department of Systems Biology, Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Gary A Bradshaw
- Department of Systems Biology, Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Fleur Jochems
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, CX, Amsterdam, The Netherlands
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, CX, Amsterdam, The Netherlands
| | - Anthony Letai
- Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Systems Biology, Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Ji XM, Dong XX, Li JP, Tai GJ, Qiu S, Wei W, Silumbwe CW, Damdinjav D, Otieno JN, Li XX, Xu M. Fisetin Clears Senescent Cells Through the Pi3k-Akt-Bcl-2/Bcl-xl Pathway to Alleviate Diabetic Aortic Aging. Phytother Res 2025. [PMID: 40259678 DOI: 10.1002/ptr.8507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/18/2025] [Accepted: 03/29/2025] [Indexed: 04/23/2025]
Abstract
Vascular aging is a major contributor to age-related cardiovascular diseases (CVDs) and type 2 diabetes mellitus (T2DM) induced early arterial aging and excessive senescent cells (SCs) burden in vessels. Inhibiting cellular senescence or eliminating SCs could effectively improve aging-related CVDs. Fisetin, a flavonoid extracted from cotinus coggygria scop, has shown potential in alleviating aging by clearing SCs. This study investigated the unexplored mechanisms and efficacy of fisetin in alleviating T2DM-related aortic aging. The T2DM mouse model was induced using a high-fat diet and low-dose streptozotocin injection. Chronic fisetin treatment's protective effects against aortic aging were assessed via senescence-associated beta-galactosidase (SA-β-Gal) staining, histopathology, and vasomotor function. RNA-sequencing and western blotting identified relevant signaling pathways and protein expression. Fisetin's effects on SCs and senescence-associated secretory phenotype (SASP) factors were evaluated through cell viability, apoptosis, and co-culture assays. Docking simulations suggested fisetin as a potential Phosphoinositide 3-kinase (Pi3k) inhibitor. In vivo, chronic fisetin treatment reduced aortic SCs burden, alleviating T2DM-related and natural aortic aging. In vitro, fisetin selectively induced apoptosis of senescent endothelial cells via regulating the Pi3k-Protein Kinase B (Akt)-B-cell lymphoma (Bcl)-2/Bcl-xl pathway and suppressed SASP and its detrimental effects. Furthermore, fisetin combined with metformin therapy showed superior anti-aging effects on T2DM-related aortic aging compared to metformin monotherapy. In conclusion, chronic fisetin treatment alleviates T2DM-related aortic aging via clearing the SCs burden and abrogating the SASP factors. Fisetin combined with metformin therapy might be a potential therapeutic strategy for T2DM-related CVDs.
Collapse
Affiliation(s)
- Xiao-Man Ji
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin-Xin Dong
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jia-Peng Li
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guang-Jie Tai
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shu Qiu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wei Wei
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ceaser Wankumbu Silumbwe
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Davaadagva Damdinjav
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Joseph Nicolao Otieno
- Institute of Traditional Medicine, Muhimbili University of Health and Allied Sciencea, Dar es Salaam, Tanzania
| | - Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
23
|
Yang Z, Potenza DM, Ming XF. Is Senolytic Therapy in Cardiovascular Diseases Ready for Translation to Clinics? Biomolecules 2025; 15:545. [PMID: 40305307 PMCID: PMC12024785 DOI: 10.3390/biom15040545] [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: 03/10/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Aging is a predominant risk factor for cardiovascular diseases. There is evidence demonstrating that senescent cells not only play a significant role in organism aging but also contribute to the pathogenesis of cardiovascular diseases in younger ages. Encouraged by recent findings that the elimination of senescent cells by pharmacogenetic tools could slow down and even reverse organism aging in animal models, senolytic drugs have been developed, and the translation of results from basic research to clinical settings has been initiated. Because numerous studies in the literature show beneficial therapeutic effects of targeting senescent cells in cardiomyopathies associated with aging and ischemia/reperfusion and in atherosclerotic vascular disease, senolytic drugs are considered the next generation of therapies for cardiovascular disorders. However, recent studies have reported controversial results or detrimental effects caused by senolytic therapeutic approaches, including worsening of cardiac dysfunction, instability of atherosclerotic plaques, and even an increase in mortality in animal models, which challenges the translation of senolytic therapy into the clinical practice. This brief review article will focus on (1) analyzing and discussing the beneficial and detrimental effects of senolytic therapeutic approaches in cardiovascular diseases and cardiovascular aging and (2) future research directions and questions that are essential to understand the controversies and to translate preclinical results of senolytic therapies into clinical practice.
Collapse
Affiliation(s)
- Zhihong Yang
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland; (D.M.P.); (X.-F.M.)
| | | | | |
Collapse
|
24
|
Yao GS, Dai JS, Fu LM, Lin J, Tan ZP, Dai L, Chen W, Luo JH, Wei JH. Development and validation of hierarchical signature for precision individualized therapy based on the landscape associated with necroptosis in clear cell renal cell carcinoma. Front Pharmacol 2025; 16:1470145. [PMID: 40255563 PMCID: PMC12006085 DOI: 10.3389/fphar.2025.1470145] [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: 07/25/2024] [Accepted: 03/04/2025] [Indexed: 04/22/2025] Open
Abstract
Background Increasing evidence is showing that necroptosis has unique clinical significance in the occurrence and development of multiple diseases. Here, we systematically evaluate the role of necroptosis in clear cell renal cell carcinoma (ccRCC) and analyze its regulatory patterns. Methods First, we evaluated the expression and enrichment of necroptotic factors in ccRCC using gene set enrichment analysis (GSEA) and survival analysis in the expression profile from The Cancer Genome Atlas (TCGA) to demonstrate the overall mutation of necroptotic pathway genes. Then, we used unsupervised clustering to divide the samples into two subtypes related to necroptosis with significant differences in overall survival (OS) and subsequently detected the differentially expressed genes (DEGs) between them. Based on this, we constructed the necroptosis scoring system (NSS), which also performed outstandingly in hierarchical data. Finally, we analyzed the association between NSS and clinical parameters, immune infiltration, and the efficacy of immunotherapy containing immune checkpoint inhibitors (ICIs), and we suggested potential therapeutic strategies. Results We screened 97 necroptosis-related genes and demonstrated that they were dysregulated in ccRCC. Using Cox analysis and least absolute shrinkage and selection operator (LASSO) regression, a prognostic prediction signature of seven genes was built. Receiver operating characteristic (ROC) curves and Kaplan-Meier (KM) analyses both showed that the model was accurate, and univariate/multivariate Cox analysis showed that as an independent prognostic factor, the higher the risk score, the poorer the survival outcome. Furthermore, the predicted scores based on the signature were observably associated with immune cell infiltration and the mutation of specific genes. In addition, the risk score could potentially predict patients' responsiveness to different chemotherapy regimens. Specifically, Nivolumab is more effective for patients with higher scores. Conclusion The necroptosis-related signature we constructed can accurately predict the prognosis of ccRCC patients and further provide clues for targeted, individualized therapy.
Collapse
Affiliation(s)
- Gao-Sheng Yao
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jun-Shang Dai
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
- Department of Obstetrics and Gynecology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Liang-Min Fu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
| | - Juan Lin
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhi-Ping Tan
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Lei Dai
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Wei Chen
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jun-Hang Luo
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Institute of Precision Medicine, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jin-Huan Wei
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Borri M, Jacobs ME, Carmeliet P, Rabelink TJ, Dumas SJ. Endothelial dysfunction in the aging kidney. Am J Physiol Renal Physiol 2025; 328:F542-F562. [PMID: 39933752 DOI: 10.1152/ajprenal.00287.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/07/2024] [Accepted: 02/07/2025] [Indexed: 02/13/2025] Open
Abstract
Global population aging is an escalating challenge in modern society, especially as it impairs the function of multiple organs and increases the burden of age-related diseases. The kidneys, in particular, experience function decline, reduced regenerative capacity, and increased susceptibility to injury as they age. As a result, the prevalence of chronic kidney disease (CKD) rises with aging, further contributing to the growing health burden in older populations. One of the key factors in this process is the dysfunction of specialized renal endothelial cells (RECs), which are essential for maintaining kidney health by regulating blood flow and supporting filtration, solute and water reabsorption, and vascular integrity. As the kidneys age, REC dysfunction drives vascular and microenvironmental changes, contributing to the overall decline in kidney function. In this review, we outline the structural and functional effects of aging on the kidney's macrovascular and microvascular compartments and provide a phenotypic description of the aged endothelium. We particularly focus on the molecular and metabolic rewiring driving and sustaining growth-arrested EC senescence phenotype. We finally give an overview of senotherapies acting on ECs, especially of those modulating metabolism. Given that the pathophysiological processes underlying kidney aging largely overlap with those observed in CKD, REC rejuvenation could also benefit patients with CKD. Moreover, such interventions may hold promise in improving the outcomes of aged kidney transplants. Hence, advancing our understanding of REC and kidney aging will create opportunities for innovations that could improve outcomes for both elderly individuals and patients with CKD.
Collapse
Affiliation(s)
- Mila Borri
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Marleen E Jacobs
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ton J Rabelink
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Sébastien J Dumas
- Department of Internal Medicine (Nephrology) & Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
26
|
Liu D, Liu Z, Hu Y, Xiong W, Wang D, Zeng Z. MOMP: A critical event in cell death regulation and anticancer treatment. Biochim Biophys Acta Rev Cancer 2025; 1880:189280. [PMID: 39947442 DOI: 10.1016/j.bbcan.2025.189280] [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: 11/04/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Mitochondrial outer membrane permeabilization (MOMP) refers to the increase in permeability of the mitochondrial outer membrane, allowing proteins, DNA, and other molecules to pass through the intermembrane space into the cytosol. As a crucial event in the induction of apoptosis, MOMP plays a significant role in regulating various forms of cell death, including apoptosis, ferroptosis, and pyroptosis. Importantly, MOMP is not a binary process of "all-or-nothing." Under sub-lethal stress stimuli, cells may experience a phenomenon referred to as minority MOMP (miMOMP), where only a subset of mitochondria undergo functional impairment, thereby disrupting the normal life cycle of the cell. This can lead to pathological and physiological changes such as tumor formation, cellular senescence, innate immune dysfunction, and chronic inflammation. This review focuses on the diversity of MOMP events to elucidate how varying degrees of MOMP under different stress conditions influence cell fate. Additionally, it summarizes the current research progress on novel antitumor therapeutic strategies targeting MOMP in clinical contexts.
Collapse
Affiliation(s)
- Dan Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Ziqi Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yan Hu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Dan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
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
|
32
|
Liu S, Xi Q, Li X, Liu H. Mitochondrial dysfunction and alveolar type II epithelial cell senescence: The destroyer and rescuer of idiopathic pulmonary fibrosis. Front Cell Dev Biol 2025; 13:1535601. [PMID: 40230412 PMCID: PMC11994736 DOI: 10.3389/fcell.2025.1535601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic respiratory disease with an unknown origin and complex pathogenic mechanisms. A deeper understanding of these mechanisms is essential for effective treatment. Pulmonary fibrosis is associated with the senescence of alveolar type II epithelial (ATⅡ) cells. Additionally, ATⅡ senescence can lead to a senescence-associated secretory phenotype, which affects cellular communication and disrupts lung tissue repair, contributing to the development of IPF. The role of mitochondrial dysfunction in senescence-related diseases is increasingly recognized. It can induce ATⅡ senescence through apoptosis, impaired autophagy, and disrupted energy metabolism, potentially playing a key role in IPF progression. This article explores the therapeutic potential of targeting cellular senescence and mitochondrial dysfunction, emphasizing their significant roles in IPF pathogenesis.
Collapse
Affiliation(s)
- Suqi Liu
- The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qian Xi
- Six Sections of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xuannian Li
- The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huaman Liu
- Six Sections of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
33
|
Darvesh S, Cash MK, Forrestall K, Maillet H, Sands D. Differential senolytic inhibition of normal versus Aβ-associated cholinesterases: implications in aging and Alzheimer's disease. Aging (Albany NY) 2025; 17:822-850. [PMID: 40159237 PMCID: PMC11984419 DOI: 10.18632/aging.206227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
Cellular senescence is a hallmark of aging and the age-related condition, Alzheimer's disease (AD). How senescence contributes to cholinergic and neuropathologic changes in AD remains uncertain. Furthermore, little is known about the relationship between senescence and cholinesterases (ChEs). Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are important in neurotransmission, cell cycle regulation, and AD amyloid-β (Aβ) pathology. Senolytic agents have shown therapeutic promise in AD models. Therefore, we evaluated in vitro and in silico activity of senolytics, dasatinib (1), nintedanib (2), fisetin (3), quercetin (4), GW2580 (5), and nootropic, meclofenoxate hydrochloride (6), toward AChE and BChE. As ChEs associated with AD pathology have altered biochemical properties, we also evaluated agents 1-6 in AD brain tissues. Enzyme kinetics showed agents 1, 3, 4, and 6 inhibited both ChEs, while 2 and 5 inhibited only AChE. Histochemistry showed inhibition of Aβ plaque-associated ChEs (1 and 2: both ChEs; 5: BChE; 6: AChE), but not normal neural-associated ChEs. Modeling studies showed 1-6 interacted with the same five binding locations of both ChEs, some of which may be allosteric sites. These agents may exert their beneficial effects, in part, by inhibiting ChEs associated with AD pathology and provide new avenues for development of next-generation inhibitors targeting pathology-associated ChEs.
Collapse
Affiliation(s)
- Sultan Darvesh
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department of Medicine (Geriatric Medicine and Neurology), Dalhousie University, Halifax, Nova Scotia B3H 2E1 Canada
| | - Meghan K. Cash
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Katrina Forrestall
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Hillary Maillet
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Dane Sands
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
34
|
McCarty TY, Kearney CJ. Human dermal fibroblast senescence in response to single and recurring oxidative stress. FRONTIERS IN AGING 2025; 6:1504977. [PMID: 40225319 PMCID: PMC11985536 DOI: 10.3389/fragi.2025.1504977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Introduction: Aging results in an accumulation of damaged cells, which reduces the health of tissues and their regenerative capabilities. In the skin, there are both internal and external drivers of oxidative stress that result in aging phenotypes. Oxidative stress has been used to model senescence in vitro; however, there has been a lack of research determining whether the severity of oxidative stress correlates with senescent phenotypes. Methods: In this work, we compare cellular and secretory responses to a single (500 μM hydrogen peroxide, 2 hours) or recurring dose of hydrogen peroxide (500 μM hydrogen peroxide, 2 hours + 4 × 300 μM hydrogen peroxide each 48 hours). Senescence induction was studied using markers including cell morphology, senescence-associated-beta-galactosidase, absence of apoptosis, and cell cycle inhibition genes. Next, functional studies of the effects of the signaling of these cells were completed, such as vascular potential, keratinocyte proliferation, and macrophage polarization. Results: Fibroblasts exposed to both single and recurring oxidative stress had increased total cell and nucleic area, increased senescence-associated-beta-galactosidase (SABGAL) expression, and they were able to escape apoptosis - all characteristics of senescent cells. Additionally, cells exposed to recurring oxidative stress expressed increased levels of cell cycle inhibitor genes and decreased expression of collagen-I, -III, and -IV. Cytokine profiling showed that the single stressed cells had a more inflammatory secretory profile. However, in functional assays, the recurring stressed cells had reduced vascular potential, reduced keratinocyte proliferation, and increased IL-1β gene expression in unpolarized and polarized macrophages. Discussion: The described protocol allows for the investigation of the direct effects of single and recurring oxidative stress in fibroblasts and their secretory effects on surrounding healthy cells. These results show that recurringly stressed fibroblasts represent a more intense senescent phenotype, which can be used in in vitro aging studies to understand the severity of senescent responses.
Collapse
Affiliation(s)
| | - Cathal J. Kearney
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
35
|
Lehoczki A, Menyhart O, Andrikovics H, Fekete M, Kiss C, Mikala G, Ungvari Z, Győrffy B. Prognostic impact of a senescence gene signature in multiple myeloma. GeroScience 2025:10.1007/s11357-025-01622-9. [PMID: 40133591 DOI: 10.1007/s11357-025-01622-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
Multiple myeloma (MM), an incurable malignancy of plasma cells, is predominantly an age-related disease, with the majority of cases occurring in patients over the age of 60. Cellular senescence, a fundamental biological process underlying aging, has been increasingly recognized for its critical role in developing age-related malignancies. In this study, we aimed to investigate the prognostic significance of genes implicated in the molecular mechanisms of senescence within a large cohort of MM patients. Gene expression and clinical data from 1416 MM patients were obtained from four GEO datasets (GSE24080, GSE4204, GSE57317, and GSE9782) and integrated into a unified database. The raw data were processed using MAS5 normalization, scaling adjustments, and JetSet probe selection to ensure cross-platform comparability. A curated set of senescence-associated genes, the SenMayo gene signature, was employed for subsequent analyses. The final gene signature was computed as a weighted mean expression of 122 senescence-associated genes, with weights derived from univariate hazard ratios. Prognostic significance was evaluated using Cox regression, Kaplan-Meier survival analysis, and multivariate models incorporating clinical parameters such as gender, isotype, and molecular subtypes. False discovery rate (FDR) correction was applied to ensure the statistical robustness of findings. The weighted SenMayo gene signature strongly correlated with overall survival in MM patients (HR = 0.6, 95% CI = 0.47-0.76, p = 1.7e-05). The 75th percent probability of survival was reached at 36.1 months in the low-expression patient group, compared to 57 months in the high-expression group. Independent validation in datasets with sufficient patient numbers confirmed the prognostic value of the SenMayo signature (GSE4204: HR = 0.58, 95% CI = 0.39-0.88, p = 0.0089; GSE24080: HR = 0.61, 95% CI = 0.45-0.83, p = 0.0012; GSE57317: HR = 0.25, 95% CI = 0.08-0.77, p = 0.0095). Multivariate analyses further established the SenMayo signature as an independent prognostic factor, even when accounting for established clinical parameters such as sex and isotype. These findings underscore the robustness and independence of the SenMayo gene signature as a predictor of overall survival in multiple myeloma. This signature provides clinically valuable insights into the role of cellular senescence in disease progression.
Collapse
Affiliation(s)
- Andrea Lehoczki
- Doctoral College, Health Sciences Division, Semmelweis University, Budapest, Hungary.
- Department of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary.
| | - Otilia Menyhart
- Department of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Hungarian Research Network, Magyar Tudósok Körútja 2, 1117, Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok Körútja. 2. 1117, Budapest, Hungary
| | - Hajnalka Andrikovics
- Healthy Aging Program, Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest, National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Monika Fekete
- Healthy Aging Program, Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Csaba Kiss
- Department of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Hungarian Research Network, Magyar Tudósok Körútja 2, 1117, Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok Körútja. 2. 1117, Budapest, Hungary
| | - Gabor Mikala
- Doctoral College, Health Sciences Division, Semmelweis University, Budapest, Hungary
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College, Health Sciences Division/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Hungarian Research Network, Magyar Tudósok Körútja 2, 1117, Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok Körútja. 2. 1117, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, Pecs, 7624, Hungary
| |
Collapse
|
36
|
Zhang Z, Zhou J, Huang R, Zhuang X, Ni S. Identification of CCNB1 as a biomarker for cellular senescence in hepatocellular carcinoma: a bioinformatics and experimental validation study. Discov Oncol 2025; 16:384. [PMID: 40128499 PMCID: PMC11933616 DOI: 10.1007/s12672-025-02182-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), originating in the liver and often asymptomatic in early stages, frequently metastasises and recures post-surgery. Currently, reliable diagnostic biomarkers and therapeutic targets for HCC are lacking. This study investigates the influence of cellular senescence on HCC, employing bioinformatics analysis and in vitro experiments to identify potential biomarkers. METHODS We integrated data from GEO microarrays (GSE14520, GSE45267 and GSE64041) to analyse differentially expressed genes (DEGs) using the R package limma. WGCNA identified gene modules highly correlated to HCC. Then, ageing-highly related differentially expressed genes (AgHDEGs) were identified. Correlation analysis, GO and KEGG functional enrichment analysis, and gene co-expression network analysis further elucidated the functions of AgHDEGs. The STRING database identified hub AgHDEGs with CCNB1 subsequently evaluated for diagnostic value using ROC curve analysis. Additionally, we explored the correlation between CCNB1 and immune cells and assessed its biological functions via GSEA. Ultimately, the conclusions from bioinformatics analysis were confirmed via in vitro experiments, complemented by molecular docking simulations of gene-drug interactions. RESULTS Eight AgHDEGs (KPNA2, CCT3, CCNB1, RACGAP1, CDKN3, FEN1, MT1X and FOXM1) were identified. PPI network analysis highlighted CCNB1 as hub AgHDEGs with ROC analysis confirming its strong diagnostic potential. Analysis of immune infiltration revealed a significant correlation between CCNB1 and M0 macrophages. Subsequent studies showed CCNB1's critical role in regulating the cell cycle. Validation experiments illustrated an upregulation of CCNB1 expression in HCC, while inhibiting CCNB1 may reduce HepG2 cell proliferation by promoting cellular senescence. Moreover, molecular docking indicated CCNB1 as a potential therapeutic target. CONCLUSION Our study underscores CCNB1's potential impact on HCC senescence and progression, suggesting its candidacy as a biomarker for HCC.
Collapse
Affiliation(s)
- Zhilan Zhang
- College of Pharmacy, Anhui Medical University, Hefei, 230000, Anhui, China
| | - Jie Zhou
- College of Pharmacy, Anhui Medical University, Hefei, 230000, Anhui, China
| | - Ruiru Huang
- College of Pharmacy, Anhui Medical University, Hefei, 230000, Anhui, China
| | - Xingxing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China
| | - Shoudong Ni
- College of Pharmacy, Anhui Medical University, Hefei, 230000, Anhui, China.
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China.
| |
Collapse
|
37
|
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
|
38
|
Spiegel M. Fisetin as a Blueprint for Senotherapeutic Agents - Elucidating Geroprotective and Senolytic Properties with Molecular Modeling. Chemistry 2025; 31:e202403755. [PMID: 39688310 PMCID: PMC11914956 DOI: 10.1002/chem.202403755] [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: 10/16/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/18/2024]
Abstract
Targeting senescent cells and the factors that accelerate this pathological state has recently emerged as a novel field in medicinal chemistry. As attention shifts to synthetic substances, studies on natural agents are often overlooked. In this paper, we present a detailed computational modeling study that encompasses quantum mechanics and molecular dynamics to elucidate the senotherapeutic activity of fisetin, a natural flavonoid. The mitochondrial environment, serving as a proxy for senescence, received special attention. Throughout the study, fisetin's outstanding geroprotective properties-exhibiting significant potential against ⋅OOH, O2⋅-, and ⋅OH radicals, surpassing those of Trolox or ascorbate-were identified. Furthermore, fisetin demonstrated a high capacity to restore oxidatively damaged biomolecules to their pristine forms, thereby renewing the functionality of proteins and amino acids. The senolytic properties were examined in terms of Bcl-2 and Bcl-xL inhibition. The results indicated that fisetin not only binds effectively to these proteins but also, with appropriate modifications, may exhibit specific selectivity toward either target. This study highlights fisetin's remarkable activity in these areas and provides a molecular description of the underlying processes, paving the way for future research.
Collapse
Affiliation(s)
- Maciej Spiegel
- Department of Organic Chemistry and Pharmaceutical TechnologyFaculty of PharmacyWroclaw Medical UniversityBorowska 211A50–556WroclawPoland
| |
Collapse
|
39
|
Wei S, Le Thi P, Zhang Y, Park MY, Do K, Hoang TTT, Morgan N, Dao T, Heo J, Jo Y, Kang YJ, Cho H, Oh CM, Jang YC, Park KD, Ryu D. Hydrogen Peroxide-Releasing Hydrogel-Mediated Cellular Senescence Model for Aging Research. Biomater Res 2025; 29:0161. [PMID: 40092651 PMCID: PMC11907071 DOI: 10.34133/bmr.0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Cellular senescence, a process that induces irreversible cell cycle arrest in response to diverse stressors, is a primary contributor to aging and age-related diseases. Currently, exposure to hydrogen peroxide is a widely used technique for establishing in vitro cellular senescence models; however, this traditional method is inconsistent, laborious, and ineffective in vivo. To overcome these limitations, we have developed a hydrogen peroxide-releasing hydrogel that can readily and controllably induce senescence in conventional 2-dimensional cell cultures as well as advanced 3-dimensional microphysiological systems. Notably, we have established 2 platforms using our hydrogen peroxide-releasing hydrogel for investigating senolytics, which is a promising innovation in anti-geronic therapy. Conclusively, our advanced model presents a highly promising tool that offers a simple, versatile, convenient, effective, and highly adaptable technique for inducing cellular senescence. This innovation not only lays a crucial foundation for future research on aging but also markedly accelerates the development of novel therapeutic strategies targeting age-related diseases.
Collapse
Affiliation(s)
- Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Phuong Le Thi
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City 700000, Vietnam
| | - Yan Zhang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Moon-Young Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Khanh Do
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Thi Thai Thanh Hoang
- Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
- Atlanta VA Medical Center, Decatur, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Nyssa Morgan
- Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
- Atlanta VA Medical Center, Decatur, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Tam Dao
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jimin Heo
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - You Jung Kang
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Hansang Cho
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Young C Jang
- Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
- Atlanta VA Medical Center, Decatur, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ki-Dong Park
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| |
Collapse
|
40
|
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
|
41
|
Tang J, Li J, Hou Z, He R, Li B, Gong J, Xie Y, Meng W, Liu Y, Ouchi T, Li L, Li B. Dasatinib and Quercetin Mitigate Age-Related Alveolar Bone Inflammaging and Neutrophil Infiltration. Oral Dis 2025. [PMID: 40051132 DOI: 10.1111/odi.15291] [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: 09/13/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE Age-related alveolar bone resorption poses a major dental health challenge, yet its mechanisms and treatments are poorly understood. This study investigates the impact of dasatinib and quercetin (D + Q) treatment on senescent cells (SnCs), senescence-associated secretory phenotype (SASP), and neutrophil infiltration in aged alveolar bone, aiming to develop new strategies for combating age-related bone resorption. METHODS C57BL/6 mice (2 and 18 months) were used to examine alveolar bone resorption, inflammaging, and neutrophil infiltration. Aged mice received D + Q treatment to assess therapeutic effects. Key measurements included cementoenamel junction to the alveolar bone crest (CEJ-ABC) distance, periodontal ligament (PDL) thickness, osteometabolism markers, SnCs accumulation, SASP expression, and neutrophil infiltration. RESULTS Aged alveolar bone showed increased CEJ-ABC distance, atrophied periodontal ligament, and unbalanced osteometabolism, along with elevated SnCs, SASP, and neutrophils compared to young controls. D + Q treatment improved these conditions by reducing CEJ-ABC distance, enhancing periodontal ligament health, and boosting bone metabolism. It also lowered the expression of SnCs, SASP, and neutrophil markers. CONCLUSION D + Q treatment effectively mitigates alveolar bone aging by clearing SnCs, lowering SASP levels, and reducing neutrophil aggregation, presenting a novel approach for age-related bone resorption.
Collapse
Affiliation(s)
- Jinru Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingya Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zeyu Hou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rong He
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingzhi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiajing Gong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuhang Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wanrong Meng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunkun Liu
- Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Wang J, Shao F, Yu QX, Ye L, Wusiman D, Wu R, Tuo Z, Wang Z, Li D, Cho WC, Wei W, Feng D. The Common Hallmarks and Interconnected Pathways of Aging, Circadian Rhythms, and Cancer: Implications for Therapeutic Strategies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0612. [PMID: 40046513 PMCID: PMC11880593 DOI: 10.34133/research.0612] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/17/2025]
Abstract
The intricate relationship between cancer, circadian rhythms, and aging is increasingly recognized as a critical factor in understanding the mechanisms underlying tumorigenesis and cancer progression. Aging is a well-established primary risk factor for cancer, while disruptions in circadian rhythms are intricately associated with the tumorigenesis and progression of various tumors. Moreover, aging itself disrupts circadian rhythms, leading to physiological changes that may accelerate cancer development. Despite these connections, the specific interplay between these processes and their collective impact on cancer remains inadequately explored in the literature. In this review, we systematically explore the physiological mechanisms of circadian rhythms and their influence on cancer development. We discuss how core circadian genes impact tumor risk and prognosis, highlighting the shared hallmarks of cancer and aging such as genomic instability, cellular senescence, and chronic inflammation. Furthermore, we examine the interplay between circadian rhythms and aging, focusing on how this crosstalk contributes to tumorigenesis, tumor proliferation, and apoptosis, as well as the impact on cellular metabolism and genomic stability. By elucidating the common pathways linking aging, circadian rhythms, and cancer, this review provides new insights into the pathophysiology of cancer and identifies potential therapeutic strategies. We propose that targeting the circadian regulation of cancer hallmarks could pave the way for novel treatments, including chronotherapy and antiaging interventions, which may offer important benefits in the clinical management of cancer.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Fanglin Shao
- Department of Rehabilitation,
The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qing Xin Yu
- Department of Pathology,
Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
- Department of Pathology,
Ningbo Medical Centre Lihuili Hospital, Ningbo, Zhejiang 315040, China
| | - Luxia Ye
- Department of Public Research Platform,
Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA,
Army Medical University, Chongqing, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - William C. Cho
- Department of Clinical Oncology,
Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
- Division of Surgery and Interventional Science,
University College London, London W1W 7TS, UK
| |
Collapse
|
43
|
Miller KN, Li B, Pierce-Hoffman HR, Patel S, Lei X, Rajesh A, Teneche MG, Havas AP, Gandhi A, Macip CC, Lyu J, Victorelli SG, Woo SH, Lagnado AB, LaPorta MA, Liu T, Dasgupta N, Li S, Davis A, Korotkov A, Hultenius E, Gao Z, Altman Y, Porritt RA, Garcia G, Mogler C, Seluanov A, Gorbunova V, Kaech SM, Tian X, Dou Z, Chen C, Passos JF, Adams PD. p53 enhances DNA repair and suppresses cytoplasmic chromatin fragments and inflammation in senescent cells. Nat Commun 2025; 16:2229. [PMID: 40044657 PMCID: PMC11882782 DOI: 10.1038/s41467-025-57229-3] [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: 11/21/2024] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
Genomic instability and inflammation are distinct hallmarks of aging, but the connection between them is poorly understood. Here we report a mechanism directly linking genomic instability and inflammation in senescent cells through a mitochondria-regulated molecular circuit involving p53 and cytoplasmic chromatin fragments (CCF) that are enriched for DNA damage signaling marker γH2A.X. We show that p53 suppresses CCF accumulation and its downstream inflammatory phenotype. p53 activation suppresses CCF formation linked to enhanced DNA repair and genome integrity. Activation of p53 in aged mice by pharmacological inhibition of MDM2 reverses transcriptomic signatures of aging and age-associated accumulation of monocytes and macrophages in liver. Mitochondrial ablation in senescent cells suppresses CCF formation and activates p53 in an ATM-dependent manner, suggesting that mitochondria-dependent formation of γH2A.X + CCF dampens nuclear DNA damage signaling and p53 activity. These data provide evidence for a mitochondria-regulated p53 signaling circuit in senescent cells that controls DNA repair, genome integrity, and senescence- and age-associated inflammation, with relevance to therapeutic targeting of age-associated disease.
Collapse
Affiliation(s)
- Karl N Miller
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA.
| | - Brightany Li
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | | | - Shreeya Patel
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Xue Lei
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Adarsh Rajesh
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Marcos G Teneche
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Aaron P Havas
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Armin Gandhi
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Carolina Cano Macip
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Jun Lyu
- Laboratory of Biochemistry and Molecular Biology; National Cancer Institute; National Institutes of Health, Bethesda, MD, USA
| | - Stella G Victorelli
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Seung-Hwa Woo
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Anthony B Lagnado
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Michael A LaPorta
- NOMIS Center for Immunobiology and Microbial Pathogenesis; Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tianhui Liu
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Nirmalya Dasgupta
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
- Center for Cancer Therapy; La Jolla Institute of Immunology, La Jolla, CA, USA
| | - Sha Li
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Andrew Davis
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Anatoly Korotkov
- Departments of Biology and Medicine; University of Rochester, Rochester, NY, USA
| | - Erik Hultenius
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Zichen Gao
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Yoav Altman
- Shared Resources; NCI-designated Cancer Center; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Rebecca A Porritt
- Shared Resources; NCI-designated Cancer Center; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Guillermina Garcia
- Shared Resources; NCI-designated Cancer Center; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Carolin Mogler
- Institute of Pathology; School of Medicine and Health; Technical University Munich (TUM), Munich, Germany
| | - Andrei Seluanov
- Departments of Biology and Medicine; University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine; University of Rochester, Rochester, NY, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis; Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Xiao Tian
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Department of Medicine; Massachusetts General Research Institute, Boston, MA, USA
- Harvard Stem Cell Institute; Harvard University, Cambridge, MA, USA
| | - Chongyi Chen
- Laboratory of Biochemistry and Molecular Biology; National Cancer Institute; National Institutes of Health, Bethesda, MD, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging; Mayo Clinic, Rochester, MN, USA
| | - Peter D Adams
- Cancer Genome and Epigenetics Program; Sanford Burnham Prebys MDI, La Jolla, CA, USA.
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
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
|
47
|
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
|
48
|
Liu S, Meng Y, Zhang Y, Qiu L, Wan X, Yang X, Zhang Y, Liu X, Wen L, Lei X, Zhang B, Han J. Integrative analysis of senescence-related genes identifies robust prognostic clusters with distinct features in hepatocellular carcinoma. J Adv Res 2025; 69:107-123. [PMID: 38614215 PMCID: PMC11954806 DOI: 10.1016/j.jare.2024.04.007] [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: 11/15/2023] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024] Open
Abstract
INTRODUCTION Senescence refers to a state of permanent cell growth arrest and is regarded as a tumor suppressive mechanism, whereas accumulative evidence demonstrate that senescent cells play an adverse role during cancer progression. The scarcity of specific and reliable markers reflecting senescence level in cancer impede our understanding of this biological basis. OBJECTIVES Senescence-related genes (SRGs) were collected for integrative analysis to reveal the role of senescence in hepatocellular carcinoma (HCC). METHODS Consensus clustering was used to subtype HCC based on SRGs. Several computational methods, including single sample gene set enrichment analysis (ssGSEA), fuzzy c-means algorithm, were performed. Data of drug sensitivities were utilized to screen potential therapeutic agents for different senescence patients. Additionally, we developed a method called signature-related gene analysis (SRGA) for identification of markers relevant to phenotype of interest. Experimental strategies consisting quantitative real-time PCR (qRT-PCR), β-galactosidase assay, western blot, and tumor-T cell co-culture system were used to validate the findings in vitro. RESULTS We identified three robust prognostic clusters of HCC patients with distinct survival outcome, mutational landscape, and immune features. We further extracted signature genes of senescence clusters to construct the senescence scoring system and profile senescence level in HCC at bulk and single-cell resolution. Senescence-induced stemness reprogramming was confirmed both in silico and in vitro. HCC patients with high senescence were immune suppressed and sensitive to Tozasertib and other drugs. We suggested that MAFG, PLIN3, and 4 other genes were pertinent to HCC senescence, and MAFG potentially mediated immune suppression, senescence, and stemness. CONCLUSION Our findings provide insights into the role of SRGs in patients stratification and precision medicine.
Collapse
Affiliation(s)
- Sicheng Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Meng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaguang Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaowen Wan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyang Yang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xueqin Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linda Wen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xue Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhong Han
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
49
|
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
|
50
|
Lambert M, Miquel G, Villeneuve L, Thorin-Trescases N, Thorin E. The senolytic ABT-263 improves cognitive functions in middle-aged male, but not female, atherosclerotic LDLr -/-;hApoB 100+/+ mice. GeroScience 2025:10.1007/s11357-025-01563-3. [PMID: 39982668 DOI: 10.1007/s11357-025-01563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Accumulation of cerebral senescent cells may compromise the continuum between vascular and neuronal function, leading to damage and cognitive decline. Elimination of senescent cells might therefore preserve vascular and neuronal functions. To test this hypothesis, we used male and female atherosclerotic LDLr-/-;hApoB100+/+ mice (ATX-mice), a model of vascular cognitive impairment (VCI), treated with the senolytic ABT-263 for 3 months (3- to 6-month or 9- to 12-month old). In young male ATX mice, prevention with ABT-263 improved spatial retention memory, in association with a higher endothelial sensitivity to shear stress and a higher hippocampal CD31+ endothelial cell density, lower activation of both astrocytes and glial cells. In young females, ABT-263 tended to improve delayed memory; however, atherosclerotic plaque was magnified by ABT-263, endothelial function was unaffected, hippocampal astrocyte activation increased and expression of CD31+ cells decreased. Hence, unlike in males, ABT-263 appears deleterious in young ATX females. In middle-aged males, the curative treatment improved the learning process and memory. Although no change in endothelial function was observed, the benefits of ABT-263 were associated with a decreased expression of several inflammaging markers, a higher density of CD31+ cells and a lower activation of glial cells. In middle-aged females, ABT-263 induced a surge of inflammaging markers, associated with a slower learning process. Altogether, our data demonstrate that ABT-263 differentially affects VCI, improving cognition in male while being deleterious in female ATX mice. More studies are needed to understand the mechanisms at the basis of the sexual dimorphic effects of the senolytic ABT-263.
Collapse
Affiliation(s)
- Mélanie Lambert
- Faculty of Medicine, Department of Pharmacology and Physiology, University of Montreal, Montreal, Quebec, Canada.
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada.
| | - Géraldine Miquel
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada
| | - Louis Villeneuve
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada
| | | | - Eric Thorin
- Montreal Heart Institute, Research Center, 5000 Rue Belanger, Montreal, Quebec, H1T 1C8, Canada
- Faculty of Medicine, Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|