1
|
Lavarti R, Alvarez-Diaz T, Marti K, Kar P, Raju RP. The context-dependent effect of cellular senescence: From embryogenesis and wound healing to aging. Ageing Res Rev 2025; 109:102760. [PMID: 40318767 PMCID: PMC12145239 DOI: 10.1016/j.arr.2025.102760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Aging is characterized by a steady loss of physiological integrity, leading to impaired function and increased vulnerability to death. Cell senescence is a biological process that progresses with aging and is believed to be a key driver of age-related diseases. Senescence, a hallmark of aging, also demonstrates its beneficial physiological aspects as an anti-cancer, pro-regenerative, homeostatic, and developmental mechanism. A transitory response in which the senescent cells are quickly formed and cleared may promote tissue regeneration and organismal fitness. At the same time, senescence-related secretory phenotypes associated with extended senescence can have devastating effects. The fact that the interaction between senescent cells and their surroundings is very context-dependent may also help to explain this seemingly opposing pleiotropic function. Further, mitochondrial dysfunction is an often-unappreciated hallmark of cellular senescence and figures prominently in multiple feedback loops that induce and maintain the senescent phenotype. This review summarizes the mechanism of cellular senescence and the significance of acute senescence. We concisely introduced the context-dependent role of senescent cells and SASP, aspects of mitochondrial biology altered in the senescent cells, and their impact on the senescent phenotype. Finally, we conclude with recent therapeutic advancements targeting cellular senescence, focusing on acute injuries and age-associated diseases. Collectively, these insights provide a future roadmap for the role of senescence in organismal fitness and life span extension.
Collapse
Affiliation(s)
- Rupa Lavarti
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Tatiana Alvarez-Diaz
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kyarangelie Marti
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Parmita Kar
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States.
| |
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
|
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
|
4
|
Wood C, Saltera Z, Garcia I, Nguyen M, Rios A, Oropeza J, Ugwa D, Mukherjee U, Sehar U, Reddy PH. Age-associated changes in the heart: implications for COVID-19 therapies. Aging (Albany NY) 2025; 17:206251. [PMID: 40372276 DOI: 10.18632/aging.206251] [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: 03/03/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025]
Abstract
Cardiac aging involves progressive structural, functional, cellular, and molecular changes that impair heart function. This review explores key mechanisms, including oxidative stress, mitochondrial dysfunction, impaired autophagy, and chronic low-grade inflammation. Excess reactive oxygen species (ROS) damage heart muscle cells, contributing to fibrosis and cellular aging. Mitochondrial dysfunction reduces energy production and increases oxidative stress, accelerating cardiac decline. Impaired autophagy limits the removal of damaged proteins and organelles, while inflammation activates signaling molecules that drive tissue remodeling. Gender differences reveal estrogen's protective role in premenopausal women, with men showing greater susceptibility to heart muscle dysfunction and injury. After menopause, women lose this hormonal protection, increasing their risk of cardiovascular conditions. Ethnic disparities, particularly among underserved minority populations, emphasize how social factors such as access to care, environment, and chronic stress contribute to worsening cardiovascular outcomes. The coronavirus disease pandemic has introduced further challenges by increasing the incidence of heart damage through inflammation, blood clots, and long-term heart failure, especially in older adults with existing metabolic conditions like diabetes and high blood pressure. The virus's interaction with receptors on heart and blood vessel cells, along with a weakened immune response in older adults, intensifies cardiac aging. Emerging therapies include delivery of therapeutic extracellular vesicles, immune cell modulation, and treatments targeting mitochondria. In addition, lifestyle strategies such as regular physical activity, nutritional improvements, and stress reduction remain vital to maintaining cardiac health. Understanding how these biological and social factors intersect is critical to developing targeted strategies that promote healthy aging of the heart.
Collapse
Affiliation(s)
- Colby Wood
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Zach Saltera
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Isaiah Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Michelle Nguyen
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Andres Rios
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jacqui Oropeza
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Destiny Ugwa
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
5
|
Martinez-Fernandez V, Barascu A, Teixeira MT. Life and Death without Telomerase: The Saccharomyces cerevisiae Model. Cold Spring Harb Perspect Biol 2025; 17:a041699. [PMID: 39694811 PMCID: PMC12047662 DOI: 10.1101/cshperspect.a041699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Saccharomyces cerevisiae, a model organism in telomere biology, has been instrumental in pioneering a comprehensive understanding of the molecular processes that occur in the absence of telomerase across eukaryotes. This exploration spans investigations into telomere dynamics, intracellular signaling cascades, and organelle-mediated responses, elucidating their impact on proliferative capacity, genome stability, and cellular variability. Through the lens of budding yeast, numerous sources of cellular heterogeneity have been identified, dissected, and modeled, shedding light on the risks associated with telomeric state transitions, including the evasion of senescence. Moreover, the unraveling of the intricate interplay between the nucleus and other organelles upon telomerase inactivation has provided insights into eukaryotic evolution and cellular communication networks. These contributions, akin to milestones achieved using budding yeast, such as the discovery of the cell cycle, DNA damage checkpoint mechanisms, and DNA replication and repair processes, have been of paramount significance for the telomere field. Particularly, these insights extend to understanding replicative senescence as an anticancer mechanism in humans and enhancing our understanding of eukaryotes' evolution.
Collapse
Affiliation(s)
- Veronica Martinez-Fernandez
- Sorbonne Université, CNRS, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, F-75005 Paris, France
| | - Aurélia Barascu
- Sorbonne Université, CNRS, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, F-75005 Paris, France
| | - Maria Teresa Teixeira
- Sorbonne Université, CNRS, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, F-75005 Paris, France
| |
Collapse
|
6
|
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
|
7
|
Marmisolle I, Chacón E, Mansilla S, Ruiz S, Bresque M, Martínez J, Martínez-Zamudio RI, Herbig U, Liu J, Finkel T, Escande C, Castro L, Quijano C. Oncogene-induced senescence mitochondrial metabolism and bioenergetics drive the secretory phenotype: further characterization and comparison with other senescence-inducing stimuli. Redox Biol 2025; 82:103606. [PMID: 40158257 PMCID: PMC11997345 DOI: 10.1016/j.redox.2025.103606] [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: 12/04/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Cellular senescence is characterized by proliferation arrest and a senescence-associated secretory phenotype (SASP), that plays a role in aging and the progression of various age-related diseases. Although various metabolic alterations have been reported, no consensus exists regarding mitochondrial bioenergetics. Here we compared mitochondrial metabolism of human fibroblasts after inducing senescence with different stimuli: the oxidant hydrogen peroxide (H2O2), the genotoxic doxorubicin, serial passage, or expression of the H-RASG12V oncogene (RAS). In senescence induced by H2O2, doxorubicin or serial passage a decrease in respiratory control ratio (RCR) and coupling efficiency was noted, in relation to control cells. On the contrary, oncogene-induced senescent cells had an overall increase in respiration rates, RCR, spare respiratory capacity and coupling efficiency. In oncogene-induced senescence (OIS) the increase in respiration rates was accompanied by an increase in fatty acid catabolism, AMPK activation, and a persistent DNA damage response (DDR), that were not present in senescent cells induced by either H2O2 or doxorubicin. Inhibition of AMPK reduced mitochondrial oxygen consumption and secretion of proinflammatory cytokines in OIS. Assessment of enzymes involved in acetyl-CoA metabolism in OIS showed a 3- to 7.5-fold increase in pyruvate dehydrogenase complex (PDH), a 40% inhibition of mitochondrial aconitase, increased phosphorylation and activation of ATP-citrate lyase (ACLY), and inhibition of acetyl-CoA carboxylase (ACC). There was also a significant increase in expression and nuclear levels of the deacetylase sirtuin 6 (SIRT6). These changes can influence the sub-cellular distribution of acetyl-CoA and modulate protein acetylation reactions in the cytoplasm and nuclei. In fact, ACLY inhibition reduced histone 3 acetylation (H3K9Ac) in OIS and secretion of SASP components. In summary, our data show marked heterogeneity in mitochondrial energy metabolism of senescent cells, depending on the inducing stimulus, reveal new metabolic features of oncogene-induced senescent cells and identify AMPK and ACLY as potential targets for SASP modulation.
Collapse
Affiliation(s)
- Inés Marmisolle
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Uruguay
| | - Eliana Chacón
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Uruguay
| | - Santiago Mansilla
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Uruguay; Departamento de Métodos Cuantitativos, Facultad de Medicina, Universidad de la República, Uruguay
| | - Santiago Ruiz
- Laboratorio de Patologías del Metabolismo y el Envejecimiento, Institut Pasteur de Montevideo, Uruguay
| | - Mariana Bresque
- Laboratorio de Patologías del Metabolismo y el Envejecimiento, Institut Pasteur de Montevideo, Uruguay
| | - Jennyfer Martínez
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Uruguay
| | | | - Utz Herbig
- Center for Cell Signaling, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Jie Liu
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Carlos Escande
- Laboratorio de Patologías del Metabolismo y el Envejecimiento, Institut Pasteur de Montevideo, Uruguay
| | - Laura Castro
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Uruguay
| | - Celia Quijano
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Uruguay.
| |
Collapse
|
8
|
Zhen W, Jiang X, Li E, Germanas T, Lee MJ, Luo T, Ma X, Wang C, Chen Y, Weichselbaum RR, Lin W. Transforming malignant tumors into vulnerable phenotypes via nanoscale coordination polymer mediated cell senescence and photodynamic therapy. Biomaterials 2025; 322:123355. [PMID: 40279766 DOI: 10.1016/j.biomaterials.2025.123355] [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: 01/08/2025] [Revised: 04/01/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Induction of senescence in cancer cells can thwart the proliferation of malignant tumors. Herein we report the design of AZT-P/pyro nanoscale coordination polymer particles consisting of 3-azido-2,3-dideoxythymidine monophosphate (AZT-P) in the core and photosensitizing pyro-lipid (pyro) in the shell for potent antitumor treatment. Gradual release of AZT-P in response to an acidic tumor microenvironment transforms cancer cells with unlimited proliferation capacity into senescent cells that are vulnerable to reactive oxygen species (ROS). Pyro selectively induces ROS generation and immunogenic cell death of cancer cells upon light irradiation. Co-delivery of AZT-P and pyro in a single particle prolongs their blood circulation times and enhances their accumulation in tumors. Additionally, the induction of senescence and ROS generation both contribute to the recruitment of immune cells to the tumors, resulting in an effective immune response to inhibit the growth of large subcutaneous tumors and metastatic spread of orthotopic tumors.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States; Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, IL, 60637, United States
| | - Xiaomin Jiang
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States; Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, IL, 60637, United States
| | - En Li
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States
| | - Tomas Germanas
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States
| | - Morten J Lee
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States
| | - Taokun Luo
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States
| | - Xin Ma
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States
| | - Chaoyu Wang
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States; Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, IL, 60637, United States
| | - Yimei Chen
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, IL, 60637, United States.
| | - Wenbin Lin
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, IL, 60637, United States; Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, 5758 South Maryland Avenue, Chicago, IL, 60637, United States.
| |
Collapse
|
9
|
Lai P, Liu L, Bancaro N, Troiani M, Calì B, Li Y, Chen J, Singh PK, Arzola RA, Attanasio G, Pernigoni N, Pasquini E, Mosole S, Rinaldi A, Sgrignani J, Qiu S, Song P, Li Y, Desbats MA, Ángel AR, Mestre RP, Cavalli A, Barile L, de Bono J, Alimonti A. Mitochondrial DNA released by senescent tumor cells enhances PMN-MDSC-driven immunosuppression through the cGAS-STING pathway. Immunity 2025; 58:811-825.e7. [PMID: 40203808 DOI: 10.1016/j.immuni.2025.03.005] [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: 01/11/2024] [Revised: 09/27/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025]
Abstract
Mitochondrial dysfunction is a hallmark of cellular senescence. Here, we investigated whether senescent cells release mitochondrial (mt)DNA into the extracellular space and its impact on innate immunity. We found that both primary senescent cells and tumor cells undergoing therapy-induced senescence actively released mtDNA into the extracellular environment. mtDNA released by senescent cells was packaged within extracellular vesicles and selectively transferred to polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in the tumor microenvironment. Upon uptake, extracellular mtDNA enhanced the immunosuppressive activity of PMN-MDSCs via cGAS-STING-NF-κB signaling, thereby promoting tumor progression. While STING activation directly induced NF-κB signaling, it also activated PKR-like endoplasmic reticulum kinase (PERK), which further amplified NF-κB activity, in PMN-MDSCs. mtDNA release from senescent cells was mediated by voltage-dependent anion channels (VDACs), and pharmacological inhibition of VDAC reduced extracellular mtDNA levels, reversed PMN-MDSC-driven immunosuppression, and enhanced chemotherapy efficacy in prostate cancer mouse models. These findings suggest that targeting mtDNA release could reprogram the immunosuppressive tumor microenvironment, improving therapeutic outcomes for chemotherapy-treated patients.
Collapse
Affiliation(s)
- Ping Lai
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne 1011, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Lei Liu
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Nicolò Bancaro
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Martina Troiani
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Bianca Calì
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Yuxin Li
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Jingjing Chen
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne 1011, Switzerland
| | - Prafull Kumar Singh
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Rydell Alvarez Arzola
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Giuseppe Attanasio
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Nicolò Pernigoni
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Jacopo Sgrignani
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland; Institute for Research in Biomedicine (IRB), Bellinzona 6500, Switzerland
| | - Shi Qiu
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Pan Song
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yingrui Li
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland
| | - Maria Andrea Desbats
- Veneto Institute of Molecular Medicine (VIMM), Padova 35129, Italy; Department of Medicine, Università degli Studi di Padova, Padova 35129, Italy
| | - Azucena Rendón Ángel
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland; Laboratory of Cellular and Molecular Cardiology and Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, Lugano 6900, Switzerland
| | - Ricardo Pereira Mestre
- Oncology Institute of Southern Switzerland (IOSI) Ente Ospedaliero Cantonale (EOC), Bellinzona 6500, Switzerland
| | - Andrea Cavalli
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland; Institute for Research in Biomedicine (IRB), Bellinzona 6500, Switzerland
| | - Lucio Barile
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland; Laboratory of Cellular and Molecular Cardiology and Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, Lugano 6900, Switzerland
| | - Johann de Bono
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Bellinzona 6500, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6962, Switzerland; Veneto Institute of Molecular Medicine (VIMM), Padova 35129, Italy; Department of Medicine, Università degli Studi di Padova, Padova 35129, Italy; Oncology Institute of Southern Switzerland (IOSI) Ente Ospedaliero Cantonale (EOC), Bellinzona 6500, Switzerland; Department of Health Sciences and Technology (D-HEST), Eidgenössische Technische Hochschule (ETH) Zurich, Zurich 8092, Switzerland.
| |
Collapse
|
10
|
Rana KS, Marwah MK, Raja FNS, Dias I, Hindalekar YS, Al Tahan MA, Brown JE, Bellary S. The influence of senescent associated secretory phenotype on glucose homeostasis in C2C12 muscle cells: insights into potential p38 inhibitor interventions. J Recept Signal Transduct Res 2025; 45:118-127. [PMID: 40051308 DOI: 10.1080/10799893.2025.2475441] [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: 11/13/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
Increased accumulation of senescent cells with aging is associated with reduced ability of insulin-target tissues to utilize glucose, resulting in increased insulin resistance and glucotoxicity. We investigated the role of the senescent-associated secretory phenotype (SASP) within C2C12, skeletal muscle cells on glucose homeostasis and if such effects could be reduced by blocking pro-inflammatory pathways. C2C12 myotubes were treated with 40% conditioned media from senescent fibroblasts. Indirect glucose uptake and glycogen content were measured. The effect of SASP on the generation of reactive oxygen species [1] and mitochondrial function was also measured. The experiments above were repeated with a p38 inhibitor. 40% SASP treatment significantly decreased glucose utilization and glycogen storage within myotubes (p < 0.0001). 40% SASP was successful in inducing oxidative stress and increased mitochondrial density, whilst reducing membrane potential following 48 h of incubation (p < 0.0001) and blocking NF-κβ, restored glucose utilization (p < 0.01) despite the presence of SASP. Co-incubation of 40% SASP with an NF-κβ inhibitor eliminates excessive ROS production and restores mitochondrial activity to levels comparable to control treatment (p < 0.0001). This study shows changes in glucose homeostasis in senescent cells is mediated through SASP, and interventions aimed at mitigating pro-inflammatory pathways can potentially improve insulin resistance.
Collapse
Affiliation(s)
- Karan S Rana
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| | - Mandeep K Marwah
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Farah N S Raja
- School of Engineering and Physical Science, Aston University, Birmingham, UK
| | - Irundika Dias
- School of Engineering and Physical Science, Aston University, Birmingham, UK
| | | | - Mohamad Anas Al Tahan
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - James E Brown
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| | - Srikanth Bellary
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| |
Collapse
|
11
|
Jones-Weinert C, Mainz L, Karlseder J. Telomere function and regulation from mouse models to human ageing and disease. Nat Rev Mol Cell Biol 2025; 26:297-313. [PMID: 39614014 DOI: 10.1038/s41580-024-00800-5] [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: 10/16/2024] [Indexed: 12/01/2024]
Abstract
Telomeres protect the ends of chromosomes but shorten following cell division in the absence of telomerase activity. When telomeres become critically short or damaged, a DNA damage response is activated. Telomeres then become dysfunctional and trigger cellular senescence or death. Telomere shortening occurs with ageing and may contribute to associated maladies such as infertility, neurodegeneration, cancer, lung dysfunction and haematopoiesis disorders. Telomere dysfunction (sometimes without shortening) is associated with various diseases, known as telomere biology disorders (also known as telomeropathies). Telomere biology disorders include dyskeratosis congenita, Høyeraal-Hreidarsson syndrome, Coats plus syndrome and Revesz syndrome. Although mouse models have been invaluable in advancing telomere research, full recapitulation of human telomere-related diseases in mice has been challenging, owing to key differences between the species. In this Review, we discuss telomere protection, maintenance and damage. We highlight the differences between human and mouse telomere biology that may contribute to discrepancies between human diseases and mouse models. Finally, we discuss recent efforts to generate new 'humanized' mouse models to better model human telomere biology. A better understanding of the limitations of mouse telomere models will pave the road for more human-like models and further our understanding of telomere biology disorders, which will contribute towards the development of new therapies.
Collapse
Affiliation(s)
| | - Laura Mainz
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jan Karlseder
- The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
12
|
Wang Z, Zhu H, Xiong W. Metabolism and metabolomics in senescence, aging, and age-related diseases: a multiscale perspective. Front Med 2025; 19:200-225. [PMID: 39821730 DOI: 10.1007/s11684-024-1116-0] [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: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
The pursuit of healthy aging has long rendered aging and senescence captivating. Age-related ailments, such as cardiovascular diseases, diabetes, and neurodegenerative disorders, pose significant threats to individuals. Recent studies have shed light on the intricate mechanisms encompassing genetics, epigenetics, transcriptomics, and metabolomics in the processes of senescence and aging, as well as the establishment of age-related pathologies. Amidst these underlying mechanisms governing aging and related pathology metabolism assumes a pivotal role that holds promise for intervention and therapeutics. The advancements in metabolomics techniques and analysis methods have significantly propelled the study of senescence and aging, particularly with the aid of multiscale metabolomics which has facilitated the discovery of metabolic markers and therapeutic potentials. This review provides an overview of senescence and aging, emphasizing the crucial role metabolism plays in the aging process as well as age-related diseases.
Collapse
Affiliation(s)
- Ziyi Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hongying Zhu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Wei Xiong
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| |
Collapse
|
13
|
Vallet E, Raynaud C. Cytokinin signaling connects the chloroplast function with nuclear genome stability. MOLECULAR PLANT 2025:S1674-2052(25)00106-6. [PMID: 40156183 DOI: 10.1016/j.molp.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Affiliation(s)
- Elody Vallet
- Université de Strasbourg, CNRS, Institut de Biologie Moléculaire des Plantes, Strasbourg 67000, France
| | - Cécile Raynaud
- Université Paris-Saclay, CNRS, INRAE, University Evry, Institute of Plant Sciences Paris-Saclay (IPS2), 91405 Orsay, France.
| |
Collapse
|
14
|
Kadam L, Chan K, Ahuna K, Marshall N, Myatt L. Differential activation of p53-Lamin A/C and p16-RB mediated senescence pathways in trophoblast from pregnancies complicated by type A2 Gestational Diabetes Mellitus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641461. [PMID: 40093078 PMCID: PMC11908226 DOI: 10.1101/2025.03.04.641461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Gestational diabetes mellitus (GDM) increases maternal risks such as hypertension and future type 2 diabetes while also contributing to fetal complications such as large-for-gestational-age infants and stillbirth. The placenta which is crucial for fetal development, exhibits structural and functional changes in GDM, but the impact of these alterations on placental trophoblast function remains unclear. During their differentiation villous cytotrophoblast display several characteristics of senescent cells however the role of senescence pathways in placental function remains unexplored in GDM. Here we investigate whether placental senescence pathways are altered in GDM, utilizing term villous tissue and primary trophoblasts to assess molecular changes, and determined fetal sex-based differences. Our data suggest that both p21 and p16 mediated senescence pathways are activated during trophoblast differentiation and are dysregulated in GDM placenta in a sexually dimorphic manner. We also provide evidence for increased activation of p53-Lamin A/C and p16-RB pathways in trophoblast from GDM placentas. Reduced expression of p21 and its downstream effects on GCM1 expression and βhCG secretion outline how altered physiological senescence can affect trophoblast differentiation and function. This is a seminal study highlighting how placental senescence pathways are altered in pregnancies complicated by GDM.
Collapse
|
15
|
Kuk MU, Lee YH, Kim D, Lee KS, Park JH, Yoon JH, Lee YJ, So B, Kim M, Kwon HW, Byun Y, Lee KY, Park JT. Sauchinone Ameliorates Senescence Through Reducing Mitochondrial ROS Production. Antioxidants (Basel) 2025; 14:259. [PMID: 40227233 PMCID: PMC11939387 DOI: 10.3390/antiox14030259] [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: 12/27/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
One of the major causes of senescence is oxidative stress caused by ROS, which is mainly generated from dysfunctional mitochondria. Strategies to limit mitochondrial ROS production are considered important for reversing senescence, but effective approaches to reduce them have not yet been developed. In this study, we screened the secondary metabolites that plants produce under oxidative stress and discovered sauchinone as a potential candidate. Sauchinone induced mitochondrial function recovery, enabling efficient electron transport within the electron transport chain (ETC). This led to a decrease in ROS production, a byproduct of inefficient electron transport. The reduction in ROS by sauchinone rejuvenated senescence-associated phenotypes. To understand the underlying mechanism by which sauchinone rejuvenates senescence, we carried out RNA sequencing and found VAMP8 as a key gene. VAMP8 was downregulated by sauchinone. Knockdown of VAMP8 decreased mitochondrial ROS levels and subsequently rejuvenated mitochondrial function, which was similar to the effect of sauchinone. Taken together, these studies revealed a novel mechanism by which sauchinone reduces mitochondrial ROS production by regulating mitochondrial function and VAMP8 expression. Our results open a new avenue for aging research to control senescence by regulating mitochondrial ROS production.
Collapse
Affiliation(s)
- Myeong Uk Kuk
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Yun Haeng Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Duyeol Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Kyeong Seon Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea; (K.S.L.); (Y.B.)
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Ji Ho Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Jee Hee Yoon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Yoo Jin Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Byeonghyeon So
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Minseon Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
| | - Hyung Wook Kwon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea; (K.S.L.); (Y.B.)
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Ki Yong Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea; (K.S.L.); (Y.B.)
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (Y.H.L.); (D.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (B.S.); (M.K.); (H.W.K.)
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
16
|
Yao J, Jiang Y, Zhang P, Miao Y, Wu X, Lei H, Xie Z, Tian Y, Zhao X, Li J, Zhu L, Wan M, Tang W. Genetic and pharmacological targeting of HINT2 promotes OXPHOS to alleviate inflammatory responses and cell necrosis in acute pancreatitis. Pharmacol Res 2025; 212:107620. [PMID: 39848351 DOI: 10.1016/j.phrs.2025.107620] [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: 10/24/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
The necrosis of pancreatic acinar cells is a key molecular event in the progression of acute pancreatitis (AP), with disturbances in mitochondrial energy metabolism considered to be a direct causative factor of acinar cell necrosis. Histidine triad nucleotide-binding protein 2 (HINT2) has been implicated in the development of various diseases, whereas its involvement in the progression of AP remains unclear. This study aims to investigate the role of HINT2 in AP. HINT2 expression in pancreatic tissues was significantly downregulated after AP. The results of glutathione-S-transferase (GST) pull-down and proteomics analyses revealed the involvement of HINT2 in regulating mitochondrial oxidative phosphorylation (OXPHOS) in AP mice. Moreover, lentivirus-mediated HINT2 overexpression not only alleviated AP-induced ATP depletion, but also relieved inflammatory responses and cell necrosis. Mechanistically, HINT2 interacted with cytochrome C oxidase II (MTCO2) to promote mitochondrial OXPHOS, thereby reducing ROS accumulation and inhibiting the activation of inflammatory signaling pathway. Besides, HINT2 act as a direct pharmacological target of Emo to elicit protective effects on AP. Importantly, Emo upregulates the expression of HINT2 and OXPHOS complex proteins and enhances the interaction between HINT2 and MTCO2. Furthermore, CRISPR/Cas9-mediated HINT2 knockout significantly impaired the protective effects of Emo against AP-induced mitochondrial energy metabolism disorders, inflammatory responses, and acinar cell necrosis. Overall, these results uncover a previously unexplored role for HINT2 in maintaining mitochondrial energy metabolism in pancreatic acinar cells and reveals novel mechanism and target for Emo-mediated AP remission.
Collapse
Affiliation(s)
- Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yuhong Jiang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Pengcheng Zhang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yifan Miao
- Department of Emergency Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610075, China
| | - Xiajia Wu
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hang Lei
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zhijun Xie
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yong Tian
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Xianlin Zhao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Juan Li
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Lv Zhu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China; Digestive Department, The First People's Hospital of Shuangliu District, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
17
|
Rat A, Martinez Fernandez V, Doumic M, Teixeira MT, Xu Z. Mathematical model linking telomeres to senescence in Saccharomyces cerevisiae reveals cell lineage versus population dynamics. Nat Commun 2025; 16:1024. [PMID: 39863614 PMCID: PMC11762778 DOI: 10.1038/s41467-025-56196-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: 01/03/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Telomere shortening ultimately causes replicative senescence. However, identifying the mechanisms driving replicative senescence in cell populations is challenging due to the heterogeneity of telomere lengths and the asynchrony of senescence onset. Here, we present a mathematical model of telomere shortening and replicative senescence in Saccharomyces cerevisiae which is quantitatively calibrated and validated using data of telomerase-deficient single cells. Simulations of yeast populations, where cells with varying proliferation capacities compete against each other, show that the distribution of telomere lengths of the initial population shapes population growth, especially through the distribution of cells' shortest telomere lengths. We also quantified how factors influencing cell viability independently of telomeres can impact senescence rates. Overall, we demonstrate a temporal evolution in the composition of senescent cell populations-from a state directly linked to critically short telomeres to a state where senescence onset becomes stochastic. This population structure may promote genome instability and facilitate senescence escape.
Collapse
Affiliation(s)
- Anaïs Rat
- Aix Marseille Univ, CNRS, I2M, Centrale Marseille, Marseille, France
- Sorbonne Université, CNRS, Université de Paris, Inria, Laboratoire Jacques-Louis Lions UMR7598, Paris, France
- Univ Brest, CNRS UMR 6205, Laboratoire de Mathématiques de Bretagne Atlantique, Brest, France
| | - Veronica Martinez Fernandez
- Sorbonne Université, CNRS, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, Paris, France
| | - Marie Doumic
- Sorbonne Université, CNRS, Université de Paris, Inria, Laboratoire Jacques-Louis Lions UMR7598, Paris, France.
- CMAP, Inria, IP Paris, Ecole polytechnique, CNRS, Palaiseau, France.
| | - Maria Teresa Teixeira
- Sorbonne Université, CNRS, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, Paris, France.
| | - Zhou Xu
- Sorbonne Université, CNRS, Laboratory of Computational and Quantitative Biology, LCQB, Paris, France
| |
Collapse
|
18
|
Iskandar M, Xiao Barbero M, Jaber M, Chen R, Gomez-Guevara R, Cruz E, Westerheide S. A Review of Telomere Attrition in Cancer and Aging: Current Molecular Insights and Future Therapeutic Approaches. Cancers (Basel) 2025; 17:257. [PMID: 39858038 PMCID: PMC11764024 DOI: 10.3390/cancers17020257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES As cells divide, telomeres shorten through a phenomenon known as telomere attrition, which leads to unavoidable senescence of cells. Unprotected DNA exponentially increases the odds of mutations, which can evolve into premature aging disorders and tumorigenesis. There has been growing academic and clinical interest in exploring this duality and developing optimal therapeutic strategies to combat telomere attrition in aging and cellular immortality in cancer. The purpose of this review is to provide an updated overview of telomere biology and therapeutic tactics to address aging and cancer. METHODS We used the Rayyan platform to review the PubMed database and examined the ClinicalTrial.gov registry to gain insight into clinical trials and their results. RESULTS Cancer cells activate telomerase or utilize alternative lengthening of telomeres to escape telomere shortening, leading to near immortality. Contrarily, normal cells experience telomeric erosion, contributing to premature aging disorders, such as Werner syndrome and Hutchinson-Gilford Progeria, and (2) aging-related diseases, such as neurodegenerative and cardiovascular diseases. CONCLUSIONS The literature presents several promising therapeutic approaches to potentially balance telomere maintenance in aging and shortening in cancer. This review highlights gaps in knowledge and points to the potential of these optimal interventions in preclinical and clinical studies to inform future research in cancer and aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandy Westerheide
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, ISA2015, Tampa, FL 33620, USA; (M.I.); (M.X.B.); (M.J.); (R.C.); (R.G.-G.); (E.C.)
| |
Collapse
|
19
|
Louka XP, Gumeni S, Trougakos IP. Studying Cellular Senescence Using the Model Organism Drosophila melanogaster. Methods Mol Biol 2025; 2906:281-299. [PMID: 40082363 DOI: 10.1007/978-1-0716-4426-3_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Cellular senescence, a complex biological process characterized by irreversible cell cycle arrest, contributes significantly to the development and progression of aging and of age-related diseases. Studying cellular senescence in vivo can be challenging due to the high heterogeneity and dynamic nature of senescent cells. Recently, Drosophila melanogaster has emerged as a powerful model organism for studying aging and cellular senescence due to its tractability and short lifespan, as well as due to the conservation of age-related genes and of key age-related pathways with mammals. Consequently, several research studies have utilized Drosophila to investigate the cellular mechanisms and pathways implicated in cellular senescence. Herein, we provide an overview of the assays that can be applied to study the different features of senescent cells in D. melanogaster tissues, highlighting the benefits of this model in aging research. We also emphasize the importance of selecting appropriate biomarkers for the identification of senescent cells, and the need for further understanding of the aging process including a more accurate identification and detection of senescent cells at the organismal level; a far more complex process as compared to single cells.
Collapse
Affiliation(s)
- Xanthippi P Louka
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
20
|
Qu X, Xie Z, Zhang J, Huang Y, Zhao R, Li N, Wang J, Chen L, Cui W, Luo X. Regulating Mitochondrial Aging via Targeting the Gut-Bone Axis in BMSCs With Oral Hydrogel Microspheres to Inhibit Bone Loss. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409936. [PMID: 39629509 DOI: 10.1002/smll.202409936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/18/2024] [Indexed: 01/30/2025]
Abstract
The gut-bone axis is a promising target for osteoporosis treatment, yet existing delivery systems lack precise targeting. Herein, an oral hydrogel microsphere system (E7-Lipo@Alg/Cs) is developed using gas microfluidic and ionic crosslinking technologies to deliver drugs to bone marrow mesenchymal stem cells (BMSCs) via the gut-bone axis, regulating mitochondrial aging. A BMSC-affine peptide is conjugated onto liposomes encapsulating Fisetin, followed by incorporation into alginate-calcium hydrogel microspheres. Chitosan is electrostatically adsorbed onto the microsphere surface, creating a core-shell structure that adheres to intestinal epithelial cells, withstands gastric acid, and facilitates targeted delivery to BMSCs through the intestinal-bone axis. In vitro, the system effectively enhances mitochondrial function and reverses BMSC aging, while in vivo studies demonstrate prolonged drug activity, restored osteogenic differentiation, and bone regeneration. RNA-seq indicates activation of the AMPK-SIRT1 pathway, reversing mitochondrial aging in BMSCs and promoting aged bone tissue regeneration. This oral hydrogel microsphere system provides a targeted and efficient strategy for regulating mitochondrial function and preventing bone loss, offering significant clinical potential for osteoporosis treatment.
Collapse
Affiliation(s)
- Xiao Qu
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Zhou Xie
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jun Zhang
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yanran Huang
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Runhan Zhao
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Ningdao Li
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Juan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Liang Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P.R. China
| | - Wenguo Cui
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xiaoji Luo
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Orthopedics, The first affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, 400060, P. R. China
| |
Collapse
|
21
|
Cakmak P, Jurmeister P, Divé I, Zeiner PS, Steinbach JP, Fenton TR, Plate KH, Czabanka M, Harter PN, Weber KJ. DNA methylation-based analysis reveals accelerated epigenetic aging in giant cell-enriched adult-type glioblastoma. Clin Epigenetics 2024; 16:179. [PMID: 39663543 PMCID: PMC11636044 DOI: 10.1186/s13148-024-01793-w] [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: 06/19/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Giant cell (gc)-enriched glioblastoma (gcGB) represents a distinct histological variant of isocitrate dehydrogenase wild-type adult-type glioblastoma with notable enlarged mono- or multinuclear tumor cells. While some studies suggest a survival advantage for gcGB patients, the underlying causes remain elusive. GcGBs are associated with TP53 mutations, and gcs were shown to accumulate DNA double-strand breaks and show deficient mitosis, potentially triggering cellular senescence programs. Epigenetic clocks have emerged as valuable tools for assessing tumor-induced age acceleration (DNAMethAgeAcc), which has lately proved itself as prognostic biomarker in glioblastoma. Our study aimed to comprehensively analyze the methylome and key metabolic proteins of gcGBs, hypothesizing that they undergo cellular aging programs compared to non-gcGBs. RESULTS A total of 310 epigenetically classified GBs, including 26 gcGBs, and nine adults with malignant gliomas allocating to pediatric high-grade glioma molecular subclasses (summarized as "pediatric GB") were included. DNAMethAgeAcc was computed by subtraction of chronological patient ages from DNA methylome-derived age estimations and its increase was associated with better survival within gcGB and non-gcGB. GcGBs were significantly more often allocated to the subgroup with increased DNAMethAgeAcc and demonstrated the highest DNAMethAgeAcc. Hypothetical senescence/aging-induced changes of the tumor microenvironment were addressed by tumor deconvolution, which was able to identify a cluster enriched for tumors with increased DNAMethAgeAcc. Key metabolic protein expression did not differ between gcGB and non-gcGB and tumor with versus without increased DNAMethAgeAcc but for elevated levels of one single mitochondrial marker, anti-mitochondrial protein MT-C02, in gcGBs. CONCLUSIONS With its sped-up epigenetic aging, gcGB presented as the epigenetic oldest GB variant in our cohort. Whereas the correlation between accelerated tumor-intrinsic epigenetic aging and cellular senescence in gcGB stays elusive, fostering epigenetic aging programs in GB might be of interest for future exploration of alternative treatment options in GB patients.
Collapse
Affiliation(s)
- Pinar Cakmak
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
| | - Philipp Jurmeister
- Ludwig Maximilians University Munich, University Hospital, Institute of Pathology, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between German Cancer Research Center (DKFZ) and University/University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Iris Divé
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
| | - Pia S Zeiner
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Department of Neurology, Frankfurt, Germany
| | - Joachim P Steinbach
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
| | - Tim R Fenton
- Somers Cancer Research, Southampton General Hospital, Southampton, UK
| | - Karl H Plate
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
| | - Marcus Czabanka
- Goethe University Frankfurt, University Hospital, Department of Neurosurgery, Frankfurt, Germany
| | - Patrick N Harter
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between German Cancer Research Center (DKFZ) and University/University Hospital, Ludwig Maximilians University Munich, Munich, Germany
- Ludwig Maximilians University Munich, University Hospital, Center for Neuropathology and Prion Research, Munich, Germany
| | - Katharina J Weber
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt, Germany.
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany.
| |
Collapse
|
22
|
Ye L, Lin D, Zhang W, Chen S, Zhen Y, Akkouche S, Liang X, Chong CM, Zhong HJ. AMBRA1 drives gastric cancer progression through regulation of tumor plasticity. Front Immunol 2024; 15:1494364. [PMID: 39720719 PMCID: PMC11666514 DOI: 10.3389/fimmu.2024.1494364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Background Stomach adenocarcinoma (STAD) is an aggressive malignancy characterized by high tumor plasticity and heterogeneity. This study investigates the role of Autophagy and Beclin 1 Regulator 1 (AMBRA1) in regulating tumor plasticity in STAD progression. Methods Combined with clinical data, the pan-cancer analysis of AMBRA1 was performed to analyze the role of AMBRA1 in STAD. Western blot, Flow Cytometry (FCM) assay, trans-well assay, wound healing assay, MTT, Reactive Oxygen Species (ROS) assay, Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR) and staining were performed to study the effects of AMBRA1 in AGS human gastric cancer cells. An AGS gastric cancer xenograft model was constructed to further verify the role of AMBRA1 in the development of STAD. Results AMBRA1 overexpression correlated with poor overall survival in STAD and was positively associated with T cell CD4+ infiltration. High AMBRA1 expression also indicated worse prognosis in patients with high cancer-associated fibroblast infiltration. AMBRA1 depletion suppressed STAD cell proliferation, migration, and invasion in vitro. Mechanistically, AMBRA1 knockdown induced G1/S cell cycle arrest and triggered cellular senescence through epigenetic alterations, including changes in H3K9me3 levels. AMBRA1 inhibition also sensitized STAD cells to chemotherapeutic agents. In vivo studies confirmed the tumor-suppressive effects of AMBRA1 loss, resulting in reduced tumor growth and increased cellular senescence. Conclusions Our findings uncover an oncogenic role for AMBRA1 in STAD. Targeting AMBRA1 may induce tumor cell senescence, apoptosis, and potentiate anti-tumor immunity, providing a rationale for developing AMBRA1-targeted precision therapies to improve clinical outcomes in STAD patients.
Collapse
Affiliation(s)
- Liuqi Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) of China, School of Pharmacy, Jinan University, Guangzhou, China
| | - Danlei Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) of China, School of Pharmacy, Jinan University, Guangzhou, China
| | - Wen Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) of China, School of Pharmacy, Jinan University, Guangzhou, China
| | - Shiji Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) of China, School of Pharmacy, Jinan University, Guangzhou, China
| | - Yumiao Zhen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) of China, School of Pharmacy, Jinan University, Guangzhou, China
| | - Sara Akkouche
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) of China, School of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaoxu Liang
- School of Arts and Science, Guangzhou Maritime University, Guangzhou, China
| | - Cheong-Meng Chong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Hai-Jing Zhong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) of China, School of Pharmacy, Jinan University, Guangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
23
|
Yousef A, Fang L, Heidari M, Kranrod J, Seubert JM. The role of CYP-sEH derived lipid mediators in regulating mitochondrial biology and cellular senescence: implications for the aging heart. Front Pharmacol 2024; 15:1486717. [PMID: 39703395 PMCID: PMC11655241 DOI: 10.3389/fphar.2024.1486717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
Cellular senescence is a condition characterized by stable, irreversible cell cycle arrest linked to the aging process. The accumulation of senescent cells in the cardiac muscle can contribute to various cardiovascular diseases (CVD). Telomere shortening, epigenetic modifications, DNA damage, mitochondrial dysfunction, and oxidative stress are known contributors to the onset of cellular senescence in the heart. The link between mitochondrial processes and cellular senescence contributed to the age-related decline in cardiac function. These include changes in mitochondrial functions and behaviours that arise from various factors, including impaired dynamics, dysregulated biogenesis, mitophagy, mitochondrial DNA (mtDNA), reduced respiratory capacity, and mitochondrial structural changes. Thus, regulation of mitochondrial biology has a role in cellular senescence and cardiac function in aging hearts. Targeting senescent cells may provide a novel therapeutic approach for treating and preventing CVD associated with aging. CYP epoxygenases metabolize N-3 and N-6 polyunsaturated fatty acids (PUFA) into epoxylipids that are readily hydrolyzed to diol products by soluble epoxide hydrolase (sEH). Increasing epoxylipids levels or inhibition of sEH has demonstrated protective effects in the aging heart. Evidence suggests they may play a role in cellular senescence by regulating mitochondria, thus reducing adverse effects of aging in the heart. In this review, we discuss how mitochondria induce cellular senescence and how epoxylipids affect the senescence process in the aged heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mobina Heidari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Joshua Kranrod
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
24
|
Kodanch SM, Mukherjee S, Prabhu NB, Kabekkodu SP, Bhat SK, Rai PS. Altered mitochondrial homeostasis on bisphenol-A exposure and its association in developing polycystic ovary syndrome: A comprehensive review. Reprod Toxicol 2024; 130:108700. [PMID: 39181417 DOI: 10.1016/j.reprotox.2024.108700] [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: 06/17/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous endocrinopathy that is known to be one of the most common reproductive pathologies observed in premenopausal women around the globe and is particularly complex as it affects various endocrine and reproductive metabolic pathways. Endocrine-disrupting chemicals (EDCs) are considered to be environmental toxicants as they have hazardous health effects on the functioning of the human endocrine system. Among various classes of EDCs, bisphenol A (BPA) has been under meticulous investigation due to its ability to alter the endocrine processes. As there is emerging evidence suggesting that BPA-induced mitochondrial homeostasis dysfunction in various pathophysiological conditions, this review aims to provide a detailed review of how various pathways associated with ovarian mitochondrial homeostasis are impaired on BPA exposure and its mirroring effects on the PCOS phenotype. BPA exposure might cause significant damage to the mitochondrial morphology and functions through the generation of reactive oxygen species (ROS) and simultaneously downregulates the total antioxidant capacity, thereby leading to oxidative stress. BPA disrupts the mitochondrial dynamics in human cells by altering the expressions of mitochondrial fission and fusion genes, increases the senescence marker proteins, along with significant alterations in the mTOR/AMPK pathway, upregulates the expression of autophagy mediating factors, and downregulates the autophagic suppressor. Furthermore, an increase in apoptosis of the ovarian granulosa cells indicates impaired folliculogenesis. As all these key features are associated with the pathogenesis of PCOS, this review can provide a better insight into the possible associations between BPA-induced dysregulation of mitochondrial homeostasis and PCOS.
Collapse
Affiliation(s)
- Supraja M Kodanch
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sayantani Mukherjee
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Navya B Prabhu
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shashikala K Bhat
- Department of Obstetrics and Gynaecology, Dr T M A Pai Hospital, Udupi, Karnataka 576101, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
25
|
Balamurli G, Liew AQX, Tee WW, Pervaiz S. Interplay between epigenetics, senescence and cellular redox metabolism in cancer and its therapeutic implications. Redox Biol 2024; 78:103441. [PMID: 39612910 PMCID: PMC11629570 DOI: 10.1016/j.redox.2024.103441] [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: 10/29/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
There is accumulating evidence indicating a close crosstalk between key molecular events regulating cell growth and proliferation, which could profoundly impact carcinogenesis and its progression. Here we focus on reviewing observations highlighting the interplay between epigenetic modifications, irreversible cell cycle arrest or senescence, and cellular redox metabolism. Epigenetic alterations, such as DNA methylation and histone modifications, dynamically influence tumour transcriptome, thereby impacting tumour phenotype, survival, growth and spread. Interestingly, the acquisition of senescent phenotype can be triggered by epigenetic changes, acting as a double-edged sword via its ability to suppress tumorigenesis or by facilitating an inflammatory milieu conducive for cancer progression. Concurrently, an aberrant redox metabolism, which is a function of the balance between reactive oxygen species (ROS) generation and intracellular anti-oxidant defences, influences signalling cascades and genomic stability in cancer cells by serving as a critical link between epigenetics and senescence. Recognizing this intricate interconnection offers a nuanced perspective for therapeutic intervention by simultaneously targeting specific epigenetic modifications, modulating senescence dynamics, and restoring redox homeostasis.
Collapse
Affiliation(s)
- Geoffrey Balamurli
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Chromatin Dynamics and Disease Epigenetics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Angeline Qiu Xia Liew
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), NUS, Singapore
| | - Wee Wei Tee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Chromatin Dynamics and Disease Epigenetics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), NUS, Singapore; NUS Medicine Healthy Longevity Program, NUS, Singapore; National University Cancer Institute, National University Health System, Singapore.
| |
Collapse
|
26
|
von Zglinicki T. Oxidative stress and cell senescence as drivers of ageing: Chicken and egg. Ageing Res Rev 2024; 102:102558. [PMID: 39454760 DOI: 10.1016/j.arr.2024.102558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Oxidative stress and cell senescence are both important drivers of ageing and age-associated disease and disability. In vitro, they are closely interconnected in a chicken-and-egg relationship: Not only is oxidative stress an important cause of cell senescence, but senescent cells are also sources of oxidative stress, obscuring cause-effect relationships during the ageing process. We hypothesize that cell senescence is a significant cause of tissue and systemic oxidative stress during ageing. This review aims to critically summarize the available evidence for this hypothesis. After summarizing the cellular feedback mechanisms that make oxidative stress an integral part of the senescent phenotype, it critically reviews the existing evidence for a role of senescent cells as causes of oxidative stress during mammalian ageing in vivo, focussing on results from intervention experiments. It is concluded that while the available data are in agreement with this hypothesis, they are still too scarce to support a robust conclusion.
Collapse
Affiliation(s)
- Thomas von Zglinicki
- Ageing Research Laboratories, Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Health, Newcastle University, UK.
| |
Collapse
|
27
|
Kuk MU, Kim D, Lee YH, Yoon JH, Park JH, Lee YJ, So BH, Kim M, Kwon HW, Byun Y, Park JT. Synergistic ROS Reduction Through the Co-Inhibition of BRAF and p38 MAPK Ameliorates Senescence. Antioxidants (Basel) 2024; 13:1465. [PMID: 39765794 PMCID: PMC11672831 DOI: 10.3390/antiox13121465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Reactive oxygen species (ROS)-mediated damage to macromolecules and cellular organelles is one of the major causes of senescence. Therapeutic strategies that lower ROS levels have been proposed as important treatments for senescence, but effective mechanisms for reducing ROS levels have not been discovered. Here, we aimed to find a combination that has a synergistic effect on ROS reduction using senomorphics known to reduce ROS. Combination treatment with BRAF inhibitor SB590885 and p38 MAPK inhibitor SB203580 showed a synergistic effect on ROS reduction compared to treatment with either drug alone. The synergistic effect of ROS reduction through this combination led to a synergistic effect that restored mitochondrial function and ameliorated senescence-associated phenotypes. To elucidate the underlying mechanism by which the synergistic effect of the two drugs reverses senescence, we performed RNA sequencing and identified metallothionein 2A (MT2A) as a key gene. MT2A was upregulated in response to combination therapy, and overexpression of MT2A led to a decrease in ROS and subsequent recovery of senescence-associated phenotypes, similar to the effects of combination therapy. Taken together, we found a drug combination that showed synergistic effects on ROS reduction, which contributed to the recovery of senescence-associated phenotypes through MT2A gene regulation. This study opens up a new avenue in aging research by demonstrating that combination therapy with existing senomorphics can enhance the ability to reverse senescence and that similar reversal effects can be achieved through gene regulation regulated by combination therapy.
Collapse
Affiliation(s)
- Myeong Uk Kuk
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Duyeol Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Yun Haeng Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Jee Hee Yoon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Ji Ho Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Yoo Jin Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Byeong Hyeon So
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Minseon Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
| | - Hyung Wook Kwon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (M.U.K.); (D.K.); (Y.H.L.); (J.H.Y.); (J.H.P.); (Y.J.L.); (B.H.S.); (M.K.); (H.W.K.)
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
28
|
Zhu J, Wu C, Yang L. Cellular senescence in Alzheimer's disease: from physiology to pathology. Transl Neurodegener 2024; 13:55. [PMID: 39568081 PMCID: PMC11577763 DOI: 10.1186/s40035-024-00447-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/12/2024] [Indexed: 11/22/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, characterized by the accumulation of Aβ and abnormal tau hyperphosphorylation. Despite substantial efforts in development of drugs targeting Aβ and tau pathologies, effective therapeutic strategies for AD remain elusive. Recent attention has been paid to the significant role of cellular senescence in AD progression. Mounting evidence suggests that interventions targeting cellular senescence hold promise in improving cognitive function and ameliorating hallmark pathologies in AD. This narrative review provides a comprehensive summary and discussion of the physiological roles, characteristics, biomarkers, and commonly employed in vivo and in vitro models of cellular senescence, with a particular focus on various cell types in the brain, including astrocytes, microglia, oligodendrocyte precursor cells, neurons, and endothelial cells. The review further delves into factors influencing cellular senescence in AD and emphasizes the significance of targeting cellular senescence as a promising approach for AD treatment, which includes the utilization of senolytics and senomorphics.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Pulmonary and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
29
|
Czaja AJ. Cellular senescence and its pathogenic and therapeutic implications in autoimmune hepatitis. Expert Rev Gastroenterol Hepatol 2024; 18:725-743. [PMID: 39575891 DOI: 10.1080/17474124.2024.2432480] [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: 09/25/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Senescent cells are characterized by replicative arrest and phenotypes that produce diverse pro-inflammatory and pro-oxidant mediators. The senescence of diverse hepatic cell types could constitute an unrecognized pathogenic mechanism and prognostic determinant in autoimmune hepatitis. The impact of cellular senescence in autoimmune hepatitis is unknown, and it may suggest adjunctive management strategies. AREAS COVERED This review describes the molecular mechanisms of cellular senescence, indicates its diagnostic features, suggests its consequences, presents possible therapeutic interventions, and encourages investigations of its pathogenic role and management in autoimmune hepatitis. Treatment prospects include elimination or reversal of senescent cells, generation of ectopic telomerase, reactivation of dormant telomerase, neutralization of specific pro-inflammatory secretory products, and mitigation of the effects of mitochondrial dysfunction. EXPERT OPINION The occurrence, nature, and consequences of cellular senescence in autoimmune hepatitis must be determined. The senescence of diverse hepatic cell types could affect the outcome of autoimmune hepatitis by impairing hepatic regeneration, intensifying liver inflammation, and worsening hepatic fibrosis. Cellular senescence could contribute to suboptimal responses during conventional glucocorticoid-based therapy. Interventions that target specific pro-inflammatory products of the senescent phenotype or selectively promote apoptosis of senescent cells may be preferred adjunctive treatments for autoimmune hepatitis depending on the cancer risk.
Collapse
Affiliation(s)
- Albert J Czaja
- Mayo Clinic, Department of Medicine, Division of Gastroenterology and Hepatology, Rochester, MN, USA
| |
Collapse
|
30
|
Cho SI, Jo ER, Jang HS. Urolithin A prevents age-related hearing loss in C57BL/6J mice likely by inducing mitophagy. Exp Gerontol 2024; 197:112589. [PMID: 39307249 DOI: 10.1016/j.exger.2024.112589] [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: 06/26/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Mitochondrial dysfunction with aging is associated with the development of age-related hearing loss. Mitophagy is a cardinal mechanism to maintain a healthy mitochondrial population through the turnover of damaged mitochondria. Declining mitophagy with age causes a buildup of damaged mitochondria, leading to sensory organ dysfunction. The effect of Urolithin A (UA), a mitophagy inducer, was investigated on age-related hearing loss in a mouse model. C57BL/6J mice were treated with UA from 6 to 10 months of age. UA attenuated an auditory brainstem responses (ABR) threshold shift at 8, 16, and 32 kHz frequencies, and improved mitochondrial DNA integrity and ATP production in the cochlea and auditory cortex. The mRNA levels of mitophagy-related genes and protein levels of PINK1, Parkin, BNIP3, and LC3B increased in the cochlea and auditory cortex. The expression of mitophagosomes and mitophagolysosomes in the cochlea, spiral ganglion, auditory cortex, and inferior colliculus increased, together with the expression of Parkin and BNIP3 in the cochlea, spiral ganglion, auditory cortex, and inferior colliculus. These results indicate that UA counteracted mitophagy decline in the auditory system and prevented age-related hearing loss. UA can be used as a potential agent to prevent age-related hearing loss.
Collapse
Affiliation(s)
- Sung Il Cho
- Department of Otolaryngology-Head and Neck Surgery, Chosun University College of Medicine, Gwangju, Republic of Korea.
| | - Eu-Ri Jo
- Department of Otolaryngology-Head and Neck Surgery, Chosun University College of Medicine, Gwangju, Republic of Korea; Department of Biomedical Sciences, Graduate School of Chosun University, Gwangju, Republic of Korea
| | - Hee Sun Jang
- Department of Otolaryngology-Head and Neck Surgery, Chosun University College of Medicine, Gwangju, Republic of Korea
| |
Collapse
|
31
|
Zhou Z, Zhang R, Zhang Y, Xu Y, Wang R, Chen S, Lv Y, Chen Y, Ren Y, Luo P, Cheng Q, Xu H, Weng S, Zuo A, Ba Y, Liu S, Han X, Liu Z. Circadian disruption in cancer hallmarks: Novel insight into the molecular mechanisms of tumorigenesis and cancer treatment. Cancer Lett 2024; 604:217273. [PMID: 39306230 DOI: 10.1016/j.canlet.2024.217273] [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: 06/27/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Circadian rhythms are 24-h rhythms governing temporal organization of behavior and physiology generated by molecular clocks composed of autoregulatory transcription-translation feedback loops (TTFLs). Disruption of circadian rhythms leads to a spectrum of pathologies, including cancer by triggering or being involved in different hallmarks. Clock control of phenotypic plasticity involved in tumorigenesis operates in aberrant dedifferentiating to progenitor-like cell states, generation of cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) events. Circadian rhythms might act as candidates for regulatory mechanisms of cellular senescent and functional determinants of senescence-associated secretory phenotype (SASP). Reciprocal control between clock and epigenetics sheds light on post-transcriptional regulation of circadian rhythms and opens avenues for novel anti-cancer strategies. Additionally, disrupting circadian rhythms influences microbiota communities that could be associated with altered homeostasis contributing to cancer development. Herein, we summarize recent advances in support of the nexus between disruptions of circadian rhythms and cancer hallmarks of new dimensions, thus providing novel perspectives on potentially effective treatment approaches for cancer management.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruiqi Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruizhi Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yingying Lv
- Department of Pediatrics, The First Affliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Pediatrics, The Third Affliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yifeng Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
32
|
Bombelli S, Grasselli C, Mazzola P, Veronesi V, Morabito I, Zucchini N, Scollo CM, Blanco SI, De Marco S, Torsello B, Vitarelli F, Antolini L, Bianchi C, Leoni V, Bellelli G, Perego RA. Impairment of Renal and Hematopoietic Stem/Progenitor Cell Compartments in Frailty Syndrome: Link With Oxidative Stress, Plasma Cytokine Profiles, and Nuclear DNA Damage. J Gerontol A Biol Sci Med Sci 2024; 79:glae188. [PMID: 39066510 PMCID: PMC11439494 DOI: 10.1093/gerona/glae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Indexed: 07/28/2024] Open
Abstract
Frailty is an age-related syndrome that drives multiple physiological system impairments in some older adults, and its pathophysiological mechanisms remain unclear. We evaluated whether frailty-related biological processes could impair stem cell compartments, specifically the renal stem compartment, given that kidney dysfunctions are frequent in frailty. A well-characterized in vitro nephrosphere model of human adult renal stem/progenitor cells has been instrumental to and was appropriate for verifying this hypothesis in our current research. Evaluating the effects of plasma from older individuals with frailty (frail plasma) on allogeneic renal stem/progenitor cells, we showed significant functional impairment and nuclear DNA damage in the treated cells of the renal stem compartment. The analysis of the frail plasma revealed mitochondrial functional impairment associated with the activation of oxidative stress and a unique inflammatory mediator profile in frail individuals. In addition, the plasma of frail subjects also contained the highest percentage of DNA-damaged autologous circulating hematopoietic progenitor/stem cells. The integration of both molecular and functional data obtained allowed us to discern patterns associated with frailty status, irrespective of the comorbidities present in the frail individuals. The data obtained converged toward biological conditions that in frailty caused renal and hematopoietic impairment of stem cells, highlighting the possibility of concomitant exhaustion of several stem compartments.
Collapse
Affiliation(s)
- Silvia Bombelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Chiara Grasselli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paolo Mazzola
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Acute Geriatric Unit, IRCCS San Gerardo, Monza, Italy
| | - Valentina Veronesi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Center - B4, University of Milano-Bicocca, Monza, Italy
| | - Ivana Morabito
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Chiara M Scollo
- Immunotransfusional Unit, Laboratory of Hematology, IRCCS San Gerardo, Monza, Italy
| | | | - Sofia De Marco
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Barbara Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Federica Vitarelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Laboratory of Clinical Pathology and Toxicology, Pio XI Hospital, ASST-Brianza, Desio, Italy
| | - Laura Antolini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Center - B4, University of Milano-Bicocca, Monza, Italy
| | - Cristina Bianchi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Valerio Leoni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Laboratory of Clinical Pathology and Toxicology, Pio XI Hospital, ASST-Brianza, Desio, Italy
| | - Giuseppe Bellelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Acute Geriatric Unit, IRCCS San Gerardo, Monza, Italy
| | - Roberto A Perego
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
33
|
Farias TG, Santos MSD, Mencalha AL, da Fonseca ADS. Low-power red laser and blue LED modulate telomere maintenance and length in human breast cancer cells. Lasers Med Sci 2024; 39:248. [PMID: 39370492 DOI: 10.1007/s10103-024-04194-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Cancer cells have the ability to undergo an unlimited number of cell divisions, which gives them immortality. Thus, the cancer cell can extend the length of its telomeres, allowing these cells to divide unlimitedly and avoid entering the state of senescence or cellular apoptosis. One of the main effects of photobiomodulation (PBM) is the increase in the production of adenosine triphosphate (ATP) and free radicals, mainly reactive oxygen species (ROS). Existent data indicates that high levels of ROS can cause shortening and dysfunctional telomeres. Therefore, a better understanding of the effects induced by PBM on cancer cell telomere maintenance is needed. This work aimed to evaluate the effects of low-power red laser (658 nm) and blue LED (470 nm) on the TRF1 and TRF2 mRNA levels and telomere length in human breast cancer cells. MCF-7 and MDA-MB-231 cells were irradiated with a low-power red laser (69 J cm-2, 0.77 W/cm-2) and blue LED (482 J cm-2, 5.35 W/cm-2), alone or in combination, and the relative mRNA levels of the genes and telomere length were assessed by quantitative reverse transcription polymerase chain reaction. The results suggested that exposure to certain red laser and blue LED fluences decreased the TRF1 and TRF2 mRNA levels in both human breast cancer cells. Telomere length was increased in MCF-7 cells after exposure to red laser and blue LED. However, telomere length in MDA-MB-231 was shortened after exposure to red laser and blue LED at fluences evaluated. Our research suggests that photobiomodulation induced by red laser and low-power blue LED could alter telomere maintenance and length.
Collapse
Affiliation(s)
- Thayssa Gomes Farias
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Boulevard Vinte e Oito de Setembro, 87, Vila Isabel, Rio de Janeiro, 20551030, Brazil.
| | - Márcia Soares Dos Santos
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Boulevard Vinte e Oito de Setembro, 87, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| | - Andre Luiz Mencalha
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Boulevard Vinte e Oito de Setembro, 87, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| | - Adenilson de Souza da Fonseca
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Boulevard Vinte e Oito de Setembro, 87, Vila Isabel, Rio de Janeiro, 20551030, Brazil
- Departamento de Ciências Fisiológicas, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rua Frei Caneca, 94, Centro, Rio de Janeiro, 20211040, Brazil
- Centro de Ciências da Saúde, Centro Universitário Serra dos Órgãos, Avenida Alberto Torres, 111, Alto, Teresópolis, Rio de Janeiro, 25964004, Brazil
| |
Collapse
|
34
|
Rubtsova MP, Nikishin DA, Vyssokikh MY, Koriagina MS, Vasiliev AV, Dontsova OA. Telomere Reprogramming and Cellular Metabolism: Is There a Link? Int J Mol Sci 2024; 25:10500. [PMID: 39408829 PMCID: PMC11476947 DOI: 10.3390/ijms251910500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Telomeres-special DNA-protein structures at the ends of linear eukaryotic chromosomes-define the proliferation potential of cells. Extremely short telomeres promote a DNA damage response and cell death to eliminate cells that may have accumulated mutations after multiple divisions. However, telomere elongation is associated with the increased proliferative potential of specific cell types, such as stem and germ cells. This elongation can be permanent in these cells and is activated temporally during immune response activation and regeneration processes. The activation of telomere lengthening mechanisms is coupled with increased proliferation and the cells' need for energy and building resources. To obtain the necessary nutrients, cells are capable of finely regulating energy production and consumption, switching between catabolic and anabolic processes. In this review, we focused on the interconnection between metabolism programs and telomere lengthening mechanisms during programmed activation of proliferation, such as in germ cell maturation, early embryonic development, neoplastic lesion growth, and immune response activation. It is generally accepted that telomere disturbance influences biological processes and promotes dysfunctionality. Here, we propose that metabolic conditions within proliferating cells should be involved in regulating telomere lengthening mechanisms, and telomere length may serve as a marker of defects in cellular functionality. We propose that it is possible to reprogram metabolism in order to regulate the telomere length and proliferative activity of cells, which may be important for the development of approaches to regeneration, immune response modulation, and cancer therapy. However, further investigations in this area are necessary to improve the understanding and manipulation of the molecular mechanisms involved in the regulation of proliferation, metabolism, and aging.
Collapse
Affiliation(s)
- Maria P. Rubtsova
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
| | - Denis A. Nikishin
- Department of Embryology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (D.A.N.); (A.V.V.)
| | - Mikhail Y. Vyssokikh
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia;
| | - Maria S. Koriagina
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
| | - Andrey V. Vasiliev
- Department of Embryology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (D.A.N.); (A.V.V.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Olga A. Dontsova
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia;
- Skolkovo Institute of Science and Technology, Center for Molecular and Cellular Biology, Moscow 121205, Russia
| |
Collapse
|
35
|
Suda M, Paul KH, Tripathi U, Minamino T, Tchkonia T, Kirkland JL. Targeting Cell Senescence and Senolytics: Novel Interventions for Age-Related Endocrine Dysfunction. Endocr Rev 2024; 45:655-675. [PMID: 38500373 PMCID: PMC11405506 DOI: 10.1210/endrev/bnae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/11/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Multiple changes occur in hormonal regulation with aging and across various endocrine organs. These changes are associated with multiple age-related disorders and diseases. A better understanding of responsible underling biological mechanisms could help in the management of multiple endocrine disorders over and above hormone replacement therapy (HRT). Cellular senescence is involved in multiple biological aging processes and pathologies common in elderly individuals. Cellular senescence, which occurs in many older individuals but also across the lifespan in association with tissue damage, acute and chronic diseases, certain drugs, and genetic syndromes, may contribute to such endocrine disorders as osteoporosis, metabolic syndrome, and type 2 diabetes mellitus. Drugs that selectively induce senescent cell removal, "senolytics,", and drugs that attenuate the tissue-destructive secretory state of certain senescent cells, "senomorphics," appear to delay the onset of or alleviate multiple diseases, including but not limited to endocrine disorders such as diabetes, complications of obesity, age-related osteoporosis, and cancers as well as atherosclerosis, chronic kidney disease, neurodegenerative disorders, and many others. More than 30 clinical trials of senolytic and senomorphic agents have already been completed, are underway, or are planned for a variety of indications. Targeting senescent cells is a novel strategy that is distinct from conventional therapies such as HRT, and thus might address unmet medical needs and can potentially amplify effects of established endocrine drug regimens, perhaps allowing for dose decreases and reducing side effects.
Collapse
Affiliation(s)
- Masayoshi Suda
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Karl H Paul
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Utkarsh Tripathi
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| | - Tamara Tchkonia
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
36
|
Qu Y, Meng B, Cai S, Yang B, He Y, Fu C, Li X, Li P, Cao Z, Mao X, Teng W, Shi S. Apoptotic metabolites ameliorate bone aging phenotypes via TCOF1/FLVCR1-mediated mitochondrial homeostasis. J Nanobiotechnology 2024; 22:549. [PMID: 39237990 PMCID: PMC11378613 DOI: 10.1186/s12951-024-02820-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024] Open
Abstract
Over 50 billion cells undergo apoptosis each day in an adult human to maintain tissue homeostasis by eliminating damaged or unwanted cells. Apoptotic deficiency can lead to age-related diseases with reduced apoptotic metabolites. However, whether apoptotic metabolism regulates aging is unclear. Here, we show that aging mice and apoptosis-deficient MRL/lpr (B6.MRL-Faslpr/J) mice exhibit decreased apoptotic levels along with increased aging phenotypes in the skeletal bones, which can be rescued by the treatment with apoptosis inducer staurosporine (STS) and stem cell-derived apoptotic vesicles (apoVs). Moreover, embryonic stem cells (ESC)-apoVs can significantly reduce senescent hallmarks and mtDNA leakage to rejuvenate aging bone marrow mesenchymal stem cells (MSCs) and ameliorate senile osteoporosis when compared to MSC-apoVs. Mechanistically, ESC-apoVs use TCOF1 to upregulate mitochondrial protein transcription, resulting in FLVCR1-mediated mitochondrial functional homeostasis. Taken together, this study reveals a previously unknown role of apoptotic metabolites in ameliorating bone aging phenotypes and the unique role of TCOF1/FLVCR1 in maintaining mitochondrial homeostasis.
Collapse
Affiliation(s)
- Yan Qu
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Bowen Meng
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Simin Cai
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Benyi Yang
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Chaoran Fu
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xiangxia Li
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Peiyi Li
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Zeyuan Cao
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Wei Teng
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
| |
Collapse
|
37
|
Kang E, Kang C, Lee YS, Lee SJV. Brief guide to senescence assays using cultured mammalian cells. Mol Cells 2024; 47:100102. [PMID: 39053732 PMCID: PMC11374973 DOI: 10.1016/j.mocell.2024.100102] [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: 06/24/2024] [Revised: 07/06/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Cellular senescence is a crucial biological process associated with organismal aging and many chronic diseases. Here, we present a brief guide to mammalian senescence assays, including the measurement of cell cycle arrest, change in cellular morphology, senescence-associated β-galactosidase (SA-β-gal) staining, and the expression of senescence-associated secretory phenotype (SASP). This work will be useful for biologists with minimum expertise in cellular senescence assays.
Collapse
Affiliation(s)
- Eunseok Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Chanhee Kang
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, South Korea
| | - Seung-Jae V Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea.
| |
Collapse
|
38
|
Pacifico F, Magni F, Leonardi A, Crescenzi E. Therapy-Induced Senescence: Novel Approaches for Markers Identification. Int J Mol Sci 2024; 25:8448. [PMID: 39126015 PMCID: PMC11313450 DOI: 10.3390/ijms25158448] [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: 06/14/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Therapy-induced senescence (TIS) represents a major cellular response to anticancer treatments. Both malignant and non-malignant cells in the tumor microenvironment undergo TIS and may be harmful for cancer patients since TIS cells develop a senescence-associated secretory phenotype (SASP) that can sustain tumor growth. The SASP also modulates anti-tumor immunity, although the immune populations involved and the final results appear to be context-dependent. In addition, senescent cancer cells are able to evade senescence growth arrest and to resume proliferation, likely contributing to relapse. So, research data suggest that TIS induction negatively affects therapy outcomes in cancer patients. In line with this, new interventions aimed at the removal of senescent cells or the reprogramming of their SASP, called senotherapy, have become attractive therapeutic options. To date, the lack of reliable, cost-effective, and easy-to-use TIS biomarkers hinders the application of recent anti-senescence therapeutic approaches in the clinic. Hence, the identification of biomarkers for the detection of TIS tumor cells and TIS non-neoplastic cells is a high priority in cancer research. In this review article, we describe the current knowledge about TIS, outline critical gaps in our knowledge, and address recent advances and novel approaches for the discovery of TIS biomarkers.
Collapse
Affiliation(s)
- Francesco Pacifico
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, CNR, Via S. Pansini 5, 80131 Naples, Italy;
| | - Fulvio Magni
- Proteomics and Metabolomics Unit, Department of Medicine and Surgery, University of Milano-Bicocca, 20854 Vedano al Lambro, Italy;
| | - Antonio Leonardi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy;
| | - Elvira Crescenzi
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, CNR, Via S. Pansini 5, 80131 Naples, Italy;
| |
Collapse
|
39
|
Zhao H, Zhao H, Ji S. A Mesenchymal stem cell Aging Framework, from Mechanisms to Strategies. Stem Cell Rev Rep 2024; 20:1420-1440. [PMID: 38727878 DOI: 10.1007/s12015-024-10732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 08/13/2024]
Abstract
Mesenchymal stem cells (MSCs) are extensively researched for therapeutic applications in tissue engineering and show significant potential for clinical use. Intrinsic or extrinsic factors causing senescence may lead to reduced proliferation, aberrant differentiation, weakened immunoregulation, and increased inflammation, ultimately limiting the potential of MSCs. It is crucial to comprehend the molecular pathways and internal processes responsible for the decline in MSC function due to senescence in order to devise innovative approaches for rejuvenating senescent MSCs and enhancing MSC treatment. We investigate the main molecular processes involved in senescence, aiming to provide a thorough understanding of senescence-related issues in MSCs. Additionally, we analyze the most recent advancements in cutting-edge approaches to combat MSC senescence based on current research. We are curious whether the aging process of stem cells results in a permanent "memory" and if cellular reprogramming may potentially revert the aging epigenome to a more youthful state.
Collapse
Affiliation(s)
- Hongqing Zhao
- Nanbu County People's Hospital, Nanchong City, 637300, Sichuan Province, China
- Jinzhou Medical University, No.82 Songpo Road, Guta District, Jinzhou, 121001, Liaoning Province, China
| | - Houming Zhao
- Graduate School of PLA Medical College, Chinese PLA General Hospital, Beijing, 100083, China
| | - Shuaifei Ji
- Graduate School of PLA Medical College, Chinese PLA General Hospital, Beijing, 100083, China.
| |
Collapse
|
40
|
Kamper RS, Nygaard H, Praeger‐Jahnsen L, Ekmann A, Ditlev SB, Schultz M, Hansen SK, Hansen P, Pressel E, Suetta C. GDF-15 is associated with sarcopenia and frailty in acutely admitted older medical patients. J Cachexia Sarcopenia Muscle 2024; 15:1549-1557. [PMID: 38890783 PMCID: PMC11294026 DOI: 10.1002/jcsm.13513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Growth differentiation factor-15 (GDF-15) has been associated with senescence, lower muscle strength, and physical performance in healthy older people. Still, it is not clear whether GDF-15 can be utilized as a biomarker of sarcopenia and frailty in the early stages of hospitalization. We investigated the association of plasma GDF-15 with sarcopenia and frailty in older, acutely admitted medical patients. METHODS The present study is based on secondary analyses of cross-sectional data from the Copenhagen PROTECT study, a prospective cohort study including 1071 patients ≥65 years of age admitted to the acute medical ward at Copenhagen University Hospital, Bispebjerg, Denmark. Muscle strength was assessed using handgrip strength, and lean mass was assessed using direct segmental multifrequency bioelectrical impedance analyses and used to clarify the potential presence of sarcopenia defined according to guidelines from the European Working Group on Sarcopenia in Older People. Frailty was evaluated using the Clinical Frailty Scale. Plasma GDF-15 was measured using electrochemiluminescence assays from Meso Scale Discovery (MSD, Rockville, MD, USA). RESULTS We included 1036 patients with completed blood samples (mean age 78.9 ± 7.8 years, 53% female). The median concentration of GDF-15 was 2669.3 pg/mL. Systemic GDF-15 was significantly higher in patients with either sarcopenia (P < 0.01) or frailty (P < 0.001) compared with patients without the conditions. Optimum cut-off points of GDF-15 relating to sarcopenia and frailty were 1541 and 2166 pg/mL, respectively. CONCLUSIONS Systemic GDF-15 was higher in acutely admitted older medical patients with sarcopenia and frailty compared with patients without. The present study defined the optimum cut-off for GDF-15, related to the presence of sarcopenia and frailty, respectively. When elevated above the derived cutoffs, GDF-15 was strongly associated with frailty and sarcopenia in both crude and fully adjusted models.
Collapse
Affiliation(s)
- Rikke S. Kamper
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Hanne Nygaard
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
- Department of Emergency MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
| | - Louis Praeger‐Jahnsen
- Copenhagen Center for Translational ResearchCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
| | - Anette Ekmann
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Sisse Bolm Ditlev
- Copenhagen Center for Translational ResearchCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
| | - Martin Schultz
- Department of GeriatricsCopenhagen University Hospital, Hvidovre and AmagerHvidovreDenmark
- Department of Clinical Medicine, Faculty of HealthUniversity of CopenhagenCopenhagenDenmark
| | - Sofie Krarup Hansen
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Pernille Hansen
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Eckart Pressel
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of HealthUniversity of CopenhagenCopenhagenDenmark
| | - Charlotte Suetta
- Department of Geriatric & Palliative MedicineCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- CopenAge, Copenhagen Center for Clinical Age ResearchUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical Medicine, Faculty of HealthUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
41
|
Gu Y, Qiu Y, Li Y, Wen W. Research progress on the regulatory mechanism of cell senescence in arsenic toxicity: a systematic review. Toxicol Res (Camb) 2024; 13:tfae136. [PMID: 39184219 PMCID: PMC11339171 DOI: 10.1093/toxres/tfae136] [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: 04/01/2024] [Revised: 06/12/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024] Open
Abstract
As an element with metalloid properties, arsenic is pervasively present in the environment and is recognized as a potent carcinogen. Consequently, the issue of human arsenic exposure has become a significant concern within the global public health sector. Numerous studies have indicated that arsenic induces cellular senescence through various mechanisms, including triggering epigenetic alterations, inducing the senescence-associated secretory phenotype (SASP), promoting telomere shortening, and causing mitochondrial dysfunction. This article collates and summarizes the latest research advancements on the involvement of cellular senescence in arsenic toxicity and explores the mechanisms of arsenic-induced toxicity. This study aims to provide new perspectives and directions for future research on arsenic toxicity and the development of prevention and treatment strategies.
Collapse
Affiliation(s)
- Yun Gu
- The School of Public Health, Dali University, Dali, China
| | - Ying Qiu
- The Second People’s Hospital of Yunnan Province, Kunming, China
- Kunming Medical University, Kunming, China
| | - Yujian Li
- The Second People’s Hospital of Yunnan Province, Kunming, China
- Kunming Medical University, Kunming, China
| | - Weihua Wen
- Yunnan Center for Disease Control and Prevention, Kunming, China
| |
Collapse
|
42
|
Liu Y, Lomeli I, Kron SJ. Therapy-Induced Cellular Senescence: Potentiating Tumor Elimination or Driving Cancer Resistance and Recurrence? Cells 2024; 13:1281. [PMID: 39120312 PMCID: PMC11312217 DOI: 10.3390/cells13151281] [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: 06/01/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Cellular senescence has been increasingly recognized as a hallmark of cancer, reflecting its association with aging and inflammation, its role as a response to deregulated proliferation and oncogenic stress, and its induction by cancer therapies. While therapy-induced senescence (TIS) has been linked to resistance, recurrence, metastasis, and normal tissue toxicity, TIS also has the potential to enhance therapy response and stimulate anti-tumor immunity. In this review, we examine the Jekyll and Hyde nature of senescent cells (SnCs), focusing on how their persistence while expressing the senescence-associated secretory phenotype (SASP) modulates the tumor microenvironment through autocrine and paracrine mechanisms. Through the SASP, SnCs can mediate both resistance and response to cancer therapies. To fulfill the unmet potential of cancer immunotherapy, we consider how SnCs may influence tumor inflammation and serve as an antigen source to potentiate anti-tumor immune response. This new perspective suggests treatment approaches based on TIS to enhance immune checkpoint blockade. Finally, we describe strategies for mitigating the detrimental effects of senescence, such as modulating the SASP or targeting SnC persistence, which may enhance the overall benefits of cancer treatment.
Collapse
Affiliation(s)
| | | | - Stephen J. Kron
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
43
|
Han MH, Kwon HS, Hwang M, Park HH, Jeong JH, Park KW, Kim EJ, Yoon SJ, Yoon B, Jang JW, Hong JY, Choi SH, Koh SH. Association between osteoporosis and the rate of telomere shortening. Aging (Albany NY) 2024; 16:11151-11161. [PMID: 39074257 PMCID: PMC11315396 DOI: 10.18632/aging.206034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
A shorter leukocyte telomere length (LTL) is reported to be associated with age-related diseases, including osteoporosis. Many studies have tried identifying the association between LTL and osteoporosis, although it remains controversial. This study aimed to determine whether osteoporosis is independently associated with LTL shortening in a prospective longitudinal cohort. The KBASE study is an independent multicenter prospective cohort in South Korea, which began in 2014. We compared the LTL values for each participant at baseline and over a 2-year follow-up period. Boxplots were used to demonstrate the differences in the change in LTL over a 2-year follow-up according to osteoporosis. Multivariable linear regression was conducted to identify whether osteoporosis is independently associated with the rate of telomere shortening. A total of 233 subjects (from 55 to 88 years) from the KBASE cohort were finally enrolled in the study. We observed that the LTL decreased by approximately 1.2 kbp over 2 years. While the LTL decreased as age increased, the rate of LTL shortening did not increase with age. Multivariable linear regression analysis indicated that only osteoporosis was independently associated with rapid LTL shortening over 2 years (B, -8.08; p = 0.038). We sought to identify an association between osteoporosis and LTL shortening in an independent prospective cohort. We found that participants with osteoporosis had significantly faster LTL shortening over 2 years than those without osteoporosis. We hope this study will help elucidate the underlying mechanisms in the relationship between LTL and osteoporosis in the future.
Collapse
Affiliation(s)
- Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri 11923, South Korea
| | - Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, Guri 11923, South Korea
| | - Mina Hwang
- Department of Neurology, Hanyang University Guri Hospital, Guri 11923, South Korea
| | - Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, Guri 11923, South Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University College of Medicine, Seoul 07804, South Korea
| | - Kyung Won Park
- Department of Neurology, Dong-A Medical Center, Dong-A University College of Medicine, Busan 49201, South Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan 49241, South Korea
| | - Soo Jin Yoon
- Department of Neurology, Eulji University Hospital, Eulji University School of Medicine, Daejeon 35233, South Korea
| | - Bora Yoon
- Department of Neurology, Konyang University College of Medicine, Daejeon 35365, Republic of Korea
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Jin Yong Hong
- Department of Neurology, Yonsei University Wonju College of Medicine, Wonju 26426, South Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University College of Medicine, Incheon 22332, South Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Guri 11923, South Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul 04763, South Korea
| |
Collapse
|
44
|
Somasundaram I, Jain SM, Blot-Chabaud M, Pathak S, Banerjee A, Rawat S, Sharma NR, Duttaroy AK. Mitochondrial dysfunction and its association with age-related disorders. Front Physiol 2024; 15:1384966. [PMID: 39015222 PMCID: PMC11250148 DOI: 10.3389/fphys.2024.1384966] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Aging is a complex process that features a functional decline in many organelles. Various factors influence the aging process, such as chromosomal abnormalities, epigenetic changes, telomere shortening, oxidative stress, and mitochondrial dysfunction. Mitochondrial dysfunction significantly impacts aging because mitochondria regulate cellular energy, oxidative balance, and calcium levels. Mitochondrial integrity is maintained by mitophagy, which helps maintain cellular homeostasis, prevents ROS production, and protects against mtDNA damage. However, increased calcium uptake and oxidative stress can disrupt mitochondrial membrane potential and permeability, leading to the apoptotic cascade. This disruption causes increased production of free radicals, leading to oxidative modification and accumulation of mitochondrial DNA mutations, which contribute to cellular dysfunction and aging. Mitochondrial dysfunction, resulting from structural and functional changes, is linked to age-related degenerative diseases. This review focuses on mitochondrial dysfunction, its implications in aging and age-related disorders, and potential anti-aging strategies through targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Indumathi Somasundaram
- Biotechnology Engineering, Kolhapur Institute of Technology’s College of Engineering, Kolhapur, India
| | - Samatha M. Jain
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | | | - Surajit Pathak
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Antara Banerjee
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Sonali Rawat
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, India
| | - Neeta Raj Sharma
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | - Asim K. Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
45
|
Chen W, Shen Z, Dong W, Huang G, Yu D, Chen W, Yan X, Yu Z. Polygonatum sibiricum polysaccharide ameliorates skeletal muscle aging via mitochondria-associated membrane-mediated calcium homeostasis regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155567. [PMID: 38579644 DOI: 10.1016/j.phymed.2024.155567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND Sarcopenia, an age-related disease, is characterized by a gradual loss of muscle mass, strength, and function. It has been linked to abnormal organelle function in myotubes, including the mitochondria and endoplasmic reticulum (ER). Recent studies revealed that mitochondria-associated membranes (MAM), the sites connecting mitochondria and the ER, may be implicated in skeletal muscle aging. In this arena, the potential of Polygonatum sibiricum polysaccharide (PSP) emerges as a beacon of hope. PSP, with its remarkable antioxidant and anti-senescence properties, is on the cusp of a therapeutic revolution, offering a promising strategy to mitigate the impacts of sarcopenia. PURPOSE The objective of this research is to explore the effects of PSP on age-related muscle dysfunction and the underlying mechanisms involved both in vivo and in vitro. METHODS In this investigation, we used in vitro experiments using D-galactose (D-gal)-induced aging in C2C12 myotubes and in vivo experiments on aged mice. Key indices were assessed, including reactive oxygen species (ROS) levels, mitochondrial function, the expression of aging-related markers, and the key proteins of mitochondria and MAM fraction. Differentially expressed genes (DEGs) related to mitochondria and ER were identified, and bioinformatic analyses were performed to explore underlying mechanisms. Muscle mass and function were determined to evaluate the quantity and quality of skeletal muscle in vivo. RESULTS PSP treatment effectively mitigated oxidative stress and mitochondrial malfunction caused by D-gal in C2C12 myotubes, preserving mitochondrial fitness and reducing MAM formation. Besides, PSP attenuated D-gal-induced increases in Ca2+ concentrations intracellularly by modulating the calcium-related proteins, which were also confirmed by gene ontology (GO) analysis of DEGs. In aged mice, PSP increased muscle mass and improved grip strength, hanging time, and other parameters while reducing ROS levels and increasing antioxidant enzyme activities in skeletal muscle tissue. CONCLUSION PSP offers protection against age-associated muscle impairments. The proposed mechanism suggests that modulation of calcium homeostasis via regulation of the MAM results in a favorable functional outcome during skeletal muscle aging. The results of this study highlight the prospect of PSP as a curative intervention for sarcopenia and affiliated pathological conditions, warranting further investigation.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Zile Shen
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Wenxi Dong
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Guowei Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Dingye Yu
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Weizhe Chen
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xialin Yan
- Department of Colorectal Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Zhen Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
46
|
Zeinoun B, Teixeira MT, Barascu A. Hog1 acts in a Mec1-independent manner to counteract oxidative stress following telomerase inactivation in Saccharomyces cerevisiae. Commun Biol 2024; 7:761. [PMID: 38909140 PMCID: PMC11193714 DOI: 10.1038/s42003-024-06464-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/18/2024] [Indexed: 06/24/2024] Open
Abstract
Replicative senescence is triggered when telomeres reach critically short length and activate permanent DNA damage checkpoint-dependent cell cycle arrest. Mitochondrial dysfunction and increase in oxidative stress are both features of replicative senescence in mammalian cells. However, how reactive oxygen species levels are controlled during senescence is elusive. Here, we show that reactive oxygen species levels increase in the telomerase-negative cells of Saccharomyces cerevisiae during replicative senescence, and that this coincides with the activation of Hog1, a mammalian p38 MAPK ortholog. Hog1 counteracts increased ROS levels during replicative senescence. While Hog1 deletion accelerates replicative senescence, we found this could stem from a reduced cell viability prior to telomerase inactivation. ROS levels also increase upon telomerase inactivation when Mec1, the yeast ortholog of ATR, is mutated, suggesting that oxidative stress is not simply a consequence of DNA damage checkpoint activation in budding yeast. We speculate that oxidative stress is a conserved hallmark of telomerase-negative eukaryote cells, and that its sources and consequences can be dissected in S. cerevisiae.
Collapse
Affiliation(s)
- Bechara Zeinoun
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France
| | - Maria Teresa Teixeira
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France.
| | - Aurélia Barascu
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France.
| |
Collapse
|
47
|
Kim D, Yoo SH, Yeon GB, Oh SS, Shin WH, Kang HC, Lee CK, Kim HW, Kim DS. Senescent Astrocytes Derived from Human Pluripotent Stem Cells Reveal Age-Related Changes and Implications for Neurodegeneration. Aging Dis 2024; 16:1709-1731. [PMID: 38913048 PMCID: PMC12096905 DOI: 10.14336/ad.2024.0089] [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: 01/24/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024] Open
Abstract
Astrocytes play a crucial role in maintaining brain homeostasis by regulating synaptic activity, providing metabolic support to neurons, and modulating immune responses in the central nervous system (CNS). During aging, astrocytes undergo senescence with various changes that affect their function and frequently lead to neurodegeneration. This study presents the first evidence of senescent astrocytes derived from human pluripotent stem cells (hPSCs). These senescent hPSC-derived astrocytes exhibited altered cellular and nuclear morphologies, along with increased expression of senescence-associated markers. Additionally, nuclear localization of NFκB, telomere shortening, and frequent signs of DNA damage were observed in these cells. Furthermore, senescent astrocytes showed defects in various critical functions necessary for maintaining a healthy CNS environment, including a reduced ability to support neuronal survival and clear neurotransmitters, synaptic debris, and toxic protein aggregates. Altered structural dynamics and reduced mitochondrial function were also observed in senescent astrocytes. Notably, treating hPSC-derived senescent astrocytes with chemicals targeting reactive oxygen species or an enzyme that regulates mitochondrial function can reverse senescence phenotypes. Thus, this study offers a valuable cellular model that can be utilized to investigate the mechanisms of brain aging and may present new avenues for discovering innovative therapeutic approaches for neurodegenerative diseases.
Collapse
Affiliation(s)
- Dongyun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea.
| | - Seo Hyun Yoo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea.
| | - Gyu-Bum Yeon
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea.
| | - Seung Soo Oh
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea.
| | - Won-Ho Shin
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Korea.
| | - Hoon-Chul Kang
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea.
| | - Cheol-Koo Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea.
| | - Hyung Wook Kim
- Department of Bio-integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Korea.
| | - Dae-Sung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea.
- Department of Pediatrics, Korea University College of Medicine, Guro Hospital, Seoul, Korea
| |
Collapse
|
48
|
Borowik AK, Lawrence MM, Peelor FF, Piekarz KM, Crosswhite A, Richardson A, Miller BF, Van Remmen H, Brown JL. Senolytic treatment does not mitigate oxidative stress-induced muscle atrophy but improves muscle force generation in CuZn superoxide dismutase knockout mice. GeroScience 2024; 46:3219-3233. [PMID: 38233728 PMCID: PMC11009189 DOI: 10.1007/s11357-024-01070-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024] Open
Abstract
Oxidative stress is associated with tissue dysfunctions that can lead to reduced health. Prior work has shown that oxidative stress contributes to both muscle atrophy and cellular senescence, which is a hallmark of aging that may drive in muscle atrophy and muscle contractile dysfunction. The purpose of the study was to test the hypothesis that cellular senescence contributes to muscle atrophy or weakness. To increase potential senescence in skeletal muscle, we used a model of oxidative stress-induced muscle frailty, the CuZn superoxide dismutase knockout (Sod1KO) mouse. We treated 6-month-old wildtype (WT) and Sod1KO mice with either vehicle or a senolytic treatment of combined dasatinib (5 mg/kg) + quercetin (50 mg/kg) (D + Q) for 3 consecutive days every 15 days. We continued treatment for 7 months and sacrificed the mice at 13 months of age. Treatment with D + Q did not preserve muscle mass, reduce NMJ fragmentation, or alter muscle protein synthesis in Sod1KO mice when compared to the vehicle-treated group. However, we observed an improvement in muscle-specific force generation in Sod1KO mice treated with D + Q when compared to Sod1KO-vehicle mice. Overall, these data suggest that reducing cellular senescence via D + Q is not sufficient to mitigate loss of muscle mass in a mouse model of oxidative stress-induced muscle frailty but may mitigate some aspects of oxidative stress-induced muscle dysfunction.
Collapse
Affiliation(s)
- Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Marcus M Lawrence
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, Utah, USA
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Katarzyna M Piekarz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Abby Crosswhite
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Arlan Richardson
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA
- Department of Biochemistry & Molecular Biology, Oklahoma University Health Science Center, Oklahoma City, OK, 73104, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA
| | - Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
49
|
Fisher JS, Adán‐Barrientos I, Kumar NR, Lancaster JN. The aged microenvironment impairs BCL6 and CD40L induction in CD4 + T follicular helper cell differentiation. Aging Cell 2024; 23:e14140. [PMID: 38481058 PMCID: PMC11296098 DOI: 10.1111/acel.14140] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 06/13/2024] Open
Abstract
Weakened germinal center responses by the aged immune system result in diminished immunity against pathogens and reduced efficacy of vaccines. Prolonged contacts between activated B cells and CD4+ T cells are crucial to germinal center formation and T follicular helper cell (Tfh) differentiation, but it is unclear how aging impacts the quality of this interaction. Peptide immunization confirmed that aged mice have decreased expansion of antigen-specific germinal center B cells and reduced antibody titers. Furthermore, aging was associated with accumulated Tfh cells, even in naïve mice. Despite increased numbers, aged Tfh had reduced expression of master transcription factor BCL6 and increased expression of the ectonucleotidase CD39. In vitro activation revealed that proliferative capacity was maintained in aged CD4+ T cells, but not the costimulatory molecule CD40L. When activated in vitro by aged antigen-presenting cells, young CD4+ naïve T cells generated reduced numbers of activated cells with upregulated CD40L. To determine the contribution of cell-extrinsic influences on antigen-specific Tfh induction, young, antigen-specific B and CD4+ T cells were adoptively transferred into aged hosts prior to peptide immunization. Transferred cells had reduced expansion and differentiation into germinal center B cell and Tfh and reduced antigen-specific antibody titers when compared to young hosts. Young CD4+ T cells transferred aged hosts differentiated into Tfh cells with reduced PD-1 and BCL6 expression, and increased CD39 expression, though they maintained their mitochondrial capacity. These results highlight the role of the lymphoid microenvironment in modulating CD4+ T cell differentiation, which contributes to impaired establishment and maintenance of germinal centers.
Collapse
Affiliation(s)
| | - Irene Adán‐Barrientos
- Immunobiology LaboratoryCentro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Naveen R. Kumar
- Department of ImmunologyMayo ClinicScottsdaleArizonaUSA
- School of Life SciencesArizona State UniversityTempeArizonaUSA
| | - Jessica N. Lancaster
- Department of ImmunologyMayo ClinicScottsdaleArizonaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of Cancer BiologyMayo ClinicScottsdaleArizonaUSA
| |
Collapse
|
50
|
Huang M, Liu M, Wang R, Man Y, Zhou H, Xu ZX, Wang Y. The crosstalk between glucose metabolism and telomerase regulation in cancer. Biomed Pharmacother 2024; 175:116643. [PMID: 38696988 DOI: 10.1016/j.biopha.2024.116643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/24/2024] [Indexed: 05/04/2024] Open
Abstract
Accumulated alterations in metabolic control provide energy and anabolic demands for enhanced cancer cell proliferation. Exemplified by the Warburg effect, changes in glucose metabolism during cancer progression are widely recognized as a characteristic of metabolic disorders. Since telomerases are a vital factor in maintaining DNA integrity and stability, any damage threatening telomerases could have a severe impact on DNA and, subsequently, whole-cell homeostasis. However, it remains unclear whether the regulation of glucose metabolism in cancer is connected to the regulation of telomerase. In this review, we present the latest insights into the crosstalk between telomerase function and glucose metabolism in cancer cells. However, at this moment this subject is not well investigated that the association is mostly indirectly regulations and few explicit regulating pathways were identified between telomerase and glucose metabolism. Therefore, the information presented in this review can provide a scientific basis for further research on the detail mechanism and the clinical application of cancer therapy, which could be valuable in improving the effectiveness of chemotherapy.
Collapse
Affiliation(s)
- Mingrui Huang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, China; The First Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Mingdi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, China
| | - Ruijia Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, China; The First Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Yifan Man
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, China; The First Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, the First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|