1
|
Münz C, Campbell GR, Esclatine A, Faure M, Labonte P, Lussignol M, Orvedahl A, Altan-Bonnet N, Bartenschlager R, Beale R, Cirone M, Espert L, Jung J, Leib D, Reggiori F, Sanyal S, Spector SA, Thiel V, Viret C, Wei Y, Wileman T, Wodrich H. Autophagy machinery as exploited by viruses. AUTOPHAGY REPORTS 2025; 4:27694127.2025.2464986. [PMID: 40201908 PMCID: PMC11921968 DOI: 10.1080/27694127.2025.2464986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
Viruses adapt and modulate cellular pathways to allow their replication in host cells. The catabolic pathway of macroautophagy, for simplicity referred to as autophagy, is no exception. In this review, we discuss anti-viral functions of both autophagy and select components of the autophagy machinery, and how viruses have evaded them. Some viruses use the membrane remodeling ability of the autophagy machinery to build their replication compartments in the cytosol or efficiently egress from cells in a non-lytic fashion. Some of the autophagy machinery components and their remodeled membranes can even be found in viral particles as envelopes or single membranes around virus packages that protect them during spreading and transmission. Therefore, studies on autophagy regulation by viral infections can reveal functions of the autophagy machinery beyond lysosomal degradation of cytosolic constituents. Furthermore, they can also pinpoint molecular interactions with which the autophagy machinery can most efficiently be manipulated, and this may be relevant to develop effective disease treatments based on autophagy modulation.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich Switzerland
| | - Grant R Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of SD, Vermillion, SD, USA
| | - Audrey Esclatine
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Patrick Labonte
- eINRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada
| | - Marion Lussignol
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anthony Orvedahl
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Bartenschlager
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division Virus-Associated Carcinogenesis, Heidelberg, Germany
- German Centre for Infection Research, Heidelberg partner site, Heidelberg, Germany
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
- Division of Medicine, University College London, London, UK
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucile Espert
- University of Montpellier, Montpellier, France
- CNRS, Institut de Recherche enInfectiologie deMontpellier (IRIM), Montpellier, France
| | - Jae Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - David Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH, USA
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, Aarhus C, Denmark
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, UK
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital, San Diego, CA, USA
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland, and Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Yu Wei
- Institut Pasteur-Theravectys Joint Laboratory, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia
- Quadram Institute Bioscience, Norwich Research Park, Norfolk, UK
| | - Harald Wodrich
- sLaboratoire de Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
2
|
Duan WL, Gu LH, Guo A, Wang XJ, Ding YY, Zhang P, Zhang BG, Li Q, Yang LX. Molecular mechanisms of programmed cell death and potential targeted pharmacotherapy in ischemic stroke (Review). Int J Mol Med 2025; 56:103. [PMID: 40341937 PMCID: PMC12081036 DOI: 10.3892/ijmm.2025.5544] [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: 01/14/2025] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Stroke poses a threat to the elderly, being the second leading cause of death and the third leading cause of disability worldwide. Ischemic stroke (IS), resulting from arterial occlusion, accounts for ~85% of all strokes. The pathophysiological processes involved in IS are intricate and complex. Currently, tissue plasminogen activator (tPA) is the only Food and Drug Administration‑approved drug for the treatment of IS. However, due to its limited administration window and the risk of symptomatic hemorrhage, tPA is applicable to only ~10% of patients with stroke. Additionally, the reperfusion process associated with thrombolytic therapy can further exacerbate damage to brain tissue. Therefore, a thorough understanding of the molecular mechanisms underlying IS‑induced injury and the identification of potential protective agents is critical for effective IS treatment. Over the past few decades, advances have been made in exploring potential protective drugs for IS. The present review summarizes the specific mechanisms of various forms of programmed cell death (PCD) induced by IS and highlights potential protective drugs targeting different PCD pathways investigated over the last decade. The present review provides a theoretical foundation for basic research and insights for the development of pharmacotherapy for IS.
Collapse
Affiliation(s)
- Wan-Li Duan
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Li-Hui Gu
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Ai Guo
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Xue-Jie Wang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yi-Yue Ding
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Peng Zhang
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Bao-Gang Zhang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Qin Li
- Rehabilitation Medicine and Health College, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Li-Xia Yang
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
3
|
Chen J, Wang S, Li T, Li W, Ke X, Ma Z, Du L, Li M. Development and characterization of endolysosomal trafficking targeting chimera degraders of α 1A-adrenergic receptor. J Adv Res 2025:S2090-1232(25)00422-9. [PMID: 40490152 DOI: 10.1016/j.jare.2025.06.014] [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: 01/04/2025] [Revised: 05/05/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025] Open
Abstract
INTRODUCTION Despite the booming targeted protein degradation technologies, degrading cell membrane proteins remains an enormous challenge. In particular, only a limited approach is appropriate for the degradation of the G protein-coupled receptor (GPCR) superfamily. It is encouraging that accelerating GPCRs' endocytosis and switching their post-endocytic fate from recycling to lysosomal degradation would represent a promising strategy for developing chemical degraders of GPCRs. OBJECTIVES This study aimed to elucidate the mechanism underlying post-endocytic sorting of internalized α1A-adrenergic receptor (α1A-AR) upon agonist stimulation and put forward a unique strategy for designing chemical degraders of GPCRs utilizing α1A-AR as an exemplary target. METHODS The protein-protein interaction (PPI) of GASP1, Beclin 2, and α1A-AR was investigated by co-immunoprecipitation and GST pull-down, and the regulatory mechanism was explored using immunofluorescence imaging and biotin protection degradation assay. By conjugating the agonistic phenylephrine moiety and a Beclin 2-recruiting moiety, ML246 with linkers, the Endolysosomal Trafficking TArgeting Chimera (ETTAC) molecules were constructed as GPCR degraders for proof-of-concept studies. RESULTS Mechanistically, the binding of Beclin 2 to GASP1 is crucial to the endolysosomal sorting and degradation of α1A-ARs. Recruiting Beclin 2 to enhance the Beclin 2-GASP1 binding, the ETTAC molecular proved to be highly efficient in reducing recycling and facilitating the degradation of α1A-AR. Furthermore, the representative ETTAC, PMA-37, effectively induces the α1A-ARs degradation in transfected and cancerous cells at the nanomole range in a GASP1 and Beclin 2-dependant manner and thus exhibits significant therapeutic effects against prostate tumor and benign prostatic hyperplasia. CONCLUSIONS Proof-of-concept studies of the ETTAC degraders for GPCR successfully elucidate the roles of post-endocytic sorting proteins and applied to directing the lysosomal degradation of α1A-ARs. Consequently, the ETTAC strategy represents a promising approach for the selective degradation of GPCRs and paves the way for future drug development.
Collapse
Affiliation(s)
- Jiwei Chen
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Tong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenhua Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xuechun Ke
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Lupei Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Tropical Biological Resources (MOE), School of Pharmaceutical Sciences, Hainan University, Haikou, Hainan 570228, China.
| |
Collapse
|
4
|
Zhu Q, McElroy R, Machhar JS, Cassel J, Zheng Z, Mansoori B, Guo H, Guo S, Pangilinan C, Liang J, Shen D, Zhang L, Liu Q, Kossenkov AV, Altieri DC, Lieberman PM, Gao SJ, Feng P, Murphy ME, Song J, Salvino JM, Liang Q, Jung JU, Liang C. Kaposi's sarcoma-associated herpesvirus induces mitochondrial fission to evade host immune responses and promote viral production. Nat Microbiol 2025:10.1038/s41564-025-02018-3. [PMID: 40404827 DOI: 10.1038/s41564-025-02018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 04/16/2025] [Indexed: 05/24/2025]
Abstract
Mitochondrial dynamics are pivotal for host immune responses upon infection, yet how viruses manipulate these processes to impair host defence and enhance viral fitness remains unclear. Here we show that Kaposi's sarcoma-associated herpesvirus (KSHV), an oncogenic virus also known as human herpesvirus 8, encodes Bcl-2 (vBcl-2), which reprogrammes mitochondrial architecture. It binds with NM23-H2, a host nucleoside diphosphate (NDP) kinase, to stimulate GTP loading of the dynamin-related protein (DRP1) GTPase, which triggers mitochondrial fission, inhibits mitochondrial antiviral signalling protein (MAVS) aggregation and impairs interferon responses in cell lines. An NM23-H2-binding-defective vBcl-2 mutant fails to evoke fission, leading to defective virion assembly due to activated MAVS-IFN signalling. Notably, we identify two key interferon-stimulated genes restricting vBcl-2-dependent virion morphogenesis. Using a high-throughput drug screening, we discover an inhibitor targeting vBcl-2-NM23-H2 interaction that blocks virion production in vitro. Our study identifies a mechanism in which KSHV manipulates mitochondrial dynamics to allow for virus assembly and shows that targeting the virus-mitochondria interface represents a potential therapeutic strategy.
Collapse
Affiliation(s)
- Qing Zhu
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert McElroy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Janvhi Suresh Machhar
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Joel Cassel
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Zihan Zheng
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Behzad Mansoori
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Hongrui Guo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sen Guo
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Christian Pangilinan
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Jinghui Liang
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Dongliang Shen
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Lu Zhang
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Qin Liu
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Andrew V Kossenkov
- Program in Genome Regulation and Cell Signaling, The Wistar Institute, Philadelphia, PA, USA
| | - Dario C Altieri
- Program in Genome Regulation and Cell Signaling, The Wistar Institute, Philadelphia, PA, USA
| | - Paul M Lieberman
- Program in Genome Regulation and Cell Signaling, The Wistar Institute, Philadelphia, PA, USA
| | - Shou-Jiang Gao
- Cancer Virology Program, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Maureen E Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Jikui Song
- Department of Biochemistry, University of California, Riverside, CA, USA
| | - Joseph M Salvino
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Qiming Liang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jae U Jung
- Department of Cancer Biology, Department of Infection Biology, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chengyu Liang
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA.
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Wang J, Fu S, Zhou Y. Research progress on the autophagy gene ATG6 in planta. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2025; 359:112577. [PMID: 40412441 DOI: 10.1016/j.plantsci.2025.112577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/19/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Autophagy is a highly conserved intracellular degradation pathway in eukaryotes. Double-membrane autophagosomes engulf damaged organelles, misfolded proteins and pathogenic microorganisms and transport them to vacuoles (in yeast and plants) or lysosomes (in animals) for degradation to maintain cellular homeostasis. As a core regulatory component of class III PI3K-I and PI3K-II complexes, ATG6 is not only involved in autophagosome formation and vesicle trafficking, but also plays an important role in plant growth, development and stress responses. This paper reviews recent progress on the structural features, molecular functions and regulatory mechanisms of plant ATG6 in response to biotic and abiotic stresses, and discusses its potential application value in future stress-resistant plant breeding.
Collapse
Affiliation(s)
- Jiajun Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University/National Citrus Engineering and Technology Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Shimin Fu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University/National Citrus Engineering and Technology Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Yan Zhou
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University/National Citrus Engineering and Technology Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China.
| |
Collapse
|
6
|
Shan M, Xu L, Yang W, Sui L, Sun P, Zhuo X, Liu S. Identification of common hub genes and construction of immune regulatory networks in aplastic anemia, myelodysplastic syndromes, and acute myeloid leukemia. Front Immunol 2025; 16:1547289. [PMID: 40406144 PMCID: PMC12095185 DOI: 10.3389/fimmu.2025.1547289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Background Aplastic anemia (AA), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML) exhibit complex pathogenic mechanisms and interrelated characteristics. We aimed to identify the common hub genes, establishing a foundation for preventing disease progression. Methods We selected relevant datasets from the Gene Expression Omnibus(GEO) database for differential gene expression, gene set enrichment, and weighted gene co-expression network analyses to identify hub genes, and then validated them. Subsequent analyses included immune infiltration analysis, single-cell sequencing, and cell communication analysis. We performed Mendelian randomization to screen inflammatory factors and immune cells. We used RT-qPCR, Enzyme - Linked Immunosorbent Assay(ELISA), and cell proliferation assays to validate the identified hub genes, their relationship with cellular communication mediators and inflammatory factors, and their impact on cellular function. Results POLG and MAP2K7 were identified as common hub genes, with low expression observed across AA, MDS, and AML. There were distinct immune differentials among these diseases, with an enhanced correlation between immune cells and hub genes as the disease progressed. Macrophage Migration Inhibitory Factor(MIF) emerged as a key mediator of cellular communication. We identified 20 regulatory pathways of immune cells and inflammatory factors across different disease stages. In vitro validation confirmed low expression of the hub genes, which were inversely correlated with MIF and inflammatory factors, though they showed no significant impact on cell proliferation or migration. Conclusions POLG and MAP2K7 demonstrate crucial roles in the progression from AA to MDS and, ultimately, to AML. These genes regulate more than 20 immune regulatory pathways through MIF-mediated communication, thereby influencing disease progression.
Collapse
Affiliation(s)
- Mingliang Shan
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Li Xu
- School of Management, Shandong Second Medical University, Weifang, China
| | - Wenzhe Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lili Sui
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Ping Sun
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Xiumei Zhuo
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Tian S, Xu H, Wu X, Ding Y, Liang L, Yin H, Zeng X, Liu Y, Zhu W. Ruthenium(II) polypyridyl complexes inhibit tumor growth through stimulating immune system to increase CD8 + T cell. Eur J Med Chem 2025; 289:117470. [PMID: 40054298 DOI: 10.1016/j.ejmech.2025.117470] [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: 12/23/2024] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025]
Abstract
In this work, we have carefully designed and synthesized two Ru(II) metal complexes: [Ru(phen)2(HMPIP)](PF6)2 (6a, where phen = 1,10-phenanthroline, HMPIP = 2-(2-hydroxy-3-methylphenyl-1H-imidazo[4,5-f][1,10]phenanthroline) and [Ru(bpy)2(HMPIP)](PF6)2 (6b, where bpy = 2,2'-bipyridine). Using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to explore the cytotoxicity of 6a and 6b towards HepG2, B16, A549, SGC-7901, HCT116 and non-cancer LO2. The complexes exhibited cytotoxicity activity against HepG2 cells. The capacity of 6a and 6b to impede the proliferation and dissemination of cancer cells was evaluated by conducting proliferation and migration experiments and 3D model. The anticancer mechanism was investigated in detail. The utilization of cycle blocking assays revealed that 6a and 6b induced a G0/G1 phase arrest in HepG2 cells. The cellular uptake experiments show that the complexes enter the cell nuclei, then escape from the cell nuclei into the cytoplasm, finally accumulate in the mitochondria. Apoptosis assays and the examination of proteins indicated that the complexes were capable of efficiently inducing apoptosis in HepG2 cells. Additionally, the potential induction of autophagy-mediated cell death was explored. The observed reduction in glutathione (GSH) levels and glutathione peroxidase 4 (GPX4) expression suggested a disruption of redox homeostasis within cancer cells, an increment in malondialdehyde (MDA) amount, together with BODIPY staining experiment, confirm that 6a and 6b can induce ferroptosis. Interestingly, in a nude mouse model, 6a showed a significant suppression of tumor growth with an inhibition rate of 63.4 %, without causing any weight loss of mice. The studies on the mechanism show that 6a causes immune cell death, increase the amount of TNF-α and IFN-γ, reduce IL-10 content, which further activates immune response to increase CD8+ T cells to prevent tumor growth. Therefore, 6a inhibits the tumor growth through stimulating the immune response to increase CD8+ T cells. In addition, the experiments in vitro show that the complexes through inhibition of PI3K/AKT/mTOR signaling pathway and intrinsic mitochondria pathway to cause cell apoptosis. These results demonstrate that Ru(II) complexes may be potent anticancer candidates for HepG2 tumor.
Collapse
Affiliation(s)
- Shuang Tian
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Haixin Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaoyu Wu
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yueyao Ding
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xiandong Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Wenrun Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
8
|
Crespo-Masip M, Goodluck HA, Kim YC, Oe Y, Roach AM, Kanoo S, Lopez N, Zhang H, Badal SS, Vallon V. ASK1 limits kidney glucose reabsorption, growth, and mid-late proximal tubule KIM-1 induction when diabetes and Western diet are combined with SGLT2 inhibition. Am J Physiol Renal Physiol 2025; 328:F662-F675. [PMID: 40152436 DOI: 10.1152/ajprenal.00031.2025] [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: 01/23/2025] [Revised: 02/18/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
Selonsertib is an apoptosis signal-regulating kinase 1 inhibitor (ASK1i) that attenuated the decline in creatinine-based estimated GFR in humans with type 2 diabetes and kidney disease but increased the rate of acute kidney injury. This study explored the individual and combined kidney effects of selonsertib and the antihyperglycemic sodium-glucose cotransporter 2 inhibitor (SGLT2i) dapagliflozin in Western diet-fed male Akita mice, a murine model of early type 1 diabetes mellitus showing signs of systemic but no kidney inflammation. ASK1i reduced elevated plasma levels of proinflammatory cytokines/chemokines (IL-6, MCP1/CCL2, KC/CXCL1, and IP-10/CXCL10) without significantly changing hyperglycemia, glomerular hyperfiltration, and albuminuria or affecting the blood glucose and glomerular hyperfiltration-lowering effect of SGLT2i. A potential sign of tubular stress, SGLT2i modestly upregulated kidney cortex transcription of proinflammatory and profibrotic genes and distal tubule injury marker Ngal. Adding ASK1i to SGLT2i lowered the transcription of many of these genes, including Ngal. However, ASK1i enhanced kidney glucose reabsorption independent of SGLT2i, and combined ASK1i + SGLT2i increased kidney weight by 30%. This was associated with and positively correlated with the upregulation of the tubular stress/injury marker KIM-1, primarily in the mid-to-late proximal tubule. Combined ASK1i + SGLT2i increased the tubular injury score but not signs of kidney inflammation or fibrosis beyond a robust increase in kidney mRNA expression of Il6, Ccl2 (Mcp1), and Timp1, associated with increased plasma IL-6 levels. The data support the hypothesis that housekeeping functions of ASK1 limit glucose reabsorption and the associated growth and cellular stress induced in the mid-to-late proximal tubule by combining hyperglycemia and Western diet with SGLT2 inhibition.NEW & NOTEWORTHY Selonsertib is an apoptosis signal-regulating kinase 1 (ASK1) inhibitor that attenuated creatinine-based eGFR decline in humans with type 2 diabetes and kidney disease but increased acute kidney injury rates. Here, we report evidence in a murine model of early type 1 diabetes mellitus that housekeeping functions of ASK1 limit glucose reabsorption and the associated growth and cellular stress induced in the mid-to-late proximal tubule by combining hyperglycemia and Western diet with SGLT2 inhibition.
Collapse
MESH Headings
- Animals
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Male
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Kidney Tubules, Proximal/enzymology
- MAP Kinase Kinase Kinase 5/metabolism
- MAP Kinase Kinase Kinase 5/antagonists & inhibitors
- Hepatitis A Virus Cellular Receptor 1/metabolism
- Diet, Western/adverse effects
- Benzhydryl Compounds/pharmacology
- Glucosides/pharmacology
- Mice, Inbred C57BL
- Diabetic Nephropathies/enzymology
- Mice
- Blood Glucose/metabolism
- Blood Glucose/drug effects
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/genetics
- Renal Reabsorption/drug effects
- Disease Models, Animal
- Glucose/metabolism
- Sodium-Glucose Transporter 2
Collapse
Affiliation(s)
- Maria Crespo-Masip
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| | - Helen A Goodluck
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| | - Young Chul Kim
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| | - Yuji Oe
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| | - Allie M Roach
- Gilead Sciences, Inc., Foster City, California, United States
| | - Sadhana Kanoo
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| | - Natalia Lopez
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| | - Haiyan Zhang
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| | - Shawn S Badal
- Gilead Sciences, Inc., Foster City, California, United States
| | - Volker Vallon
- Department of Medicine, Veterans Affairs San Diego Healthcare System, University of California San Diego, California, United States
| |
Collapse
|
9
|
Zhao X, Zhao B, Li H, Liu Y, Wang B, Li A, Zeng T, Hui HX, Sun J, Cikes D, Gheldof N, Hager J, Mi J, Laybutt DR, Deng Y, Shi Y, Neely GG, Wang Q. MTCH2 Suppresses Thermogenesis by Regulating Autophagy in Adipose Tissue. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416598. [PMID: 40051328 PMCID: PMC12061245 DOI: 10.1002/advs.202416598] [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] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/17/2025] [Indexed: 05/10/2025]
Abstract
Stimulating adipose tissue thermogenesis has emerged as a promising strategy for combating obesity, with uncoupling protein 1 (UCP1) playing a central role in this process. However, the mechanisms that suppress adipose thermogenesis and energy dissipation in obesity are not fully understood. This study identifies mitochondrial carrier homolog 2 (MTCH2), an obesity susceptibility gene, as a negative regulator of energy homeostasis across flies, rodents, and humans. Notably, adipose-specific MTCH2 depletion in mice protects against high-fat-diet (HFD)-induced obesity and metabolic disorders. Mechanistically, MTCH2 deficiency promotes energy expenditure by stimulating thermogenesis in brown adipose tissue (BAT) and browning of subcutaneous white adipose tissue (scWAT), accompanied by upregulated UCP1 protein expression, enhanced mitochondrial biogenesis, and increased lipolysis in BAT and scWAT. Using integrated RNA sequencing and proteomic analyses, this study demonstrates that MTCH2 is a key suppressor of thermogenesis by negatively regulating autophagy via Bcl-2-dependent mechanism. These findings highlight MTCH2's critical role in energy homeostasis and reveal a previously unrecognized link between MTCH2, thermogenesis, and autophagy in adipose tissue biology, positioning MTCH2 as a promising therapeutic target for obesity and related metabolic disorders. This study provides new opportunities to develop treatments that enhance energy expenditure.
Collapse
Affiliation(s)
- Xin‐Yuan Zhao
- Laboratory of Metabolism and AgingSchool of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhen518107China
| | - Ben‐Chi Zhao
- Laboratory of Metabolism and AgingSchool of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhen518107China
| | - Hui‐Lin Li
- Laboratory of Metabolism and AgingSchool of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhen518107China
| | - Ying Liu
- Laboratory of Metabolism and AgingSchool of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhen518107China
| | - Bei Wang
- Laboratory of Metabolism and AgingSchool of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhen518107China
| | - An‐Qi Li
- Laboratory of Metabolism and AgingSchool of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhen518107China
| | - Tian‐Shu Zeng
- Wuhan Union HospitalHuazhong University of Science and TechnologyWuhan430022China
| | - Hannah Xiaoyan Hui
- School of Biomedical SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Jia Sun
- Department of EndocrinologyZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Domagoj Cikes
- Institute of Physiology and PathophysiologyJohannes Kepler University LinzLinz4020Austria
| | - Nele Gheldof
- Ecole Polytechnique de Lausanne (EPFL)LausanneCH‐1015Switzerland
| | - Jorg Hager
- Nestlé Institute of Health SciencesLausanneCH‐1015Switzerland
| | - Jian‐Xun Mi
- Key Laboratory of Big Data Intelligent ComputingChongqing University of Posts and TelecommunicationsChongqing400065China
- Chongqing Key Laboratory of Image CognitionChongqing University of Posts and TelecommunicationsChongqing400065China
- College of Computer Science and TechnologyChongqing University of Posts and TelecommunicationsChongqing400065China
| | - D. Ross Laybutt
- Garvan Institute of Medical ResearchSt Vincent's Clinical SchoolUNSW SydneyDarlinghurstSydneyNSW2010Australia
| | - Yin‐Yue Deng
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107China
| | - Yan‐Chuan Shi
- Neuroendocrinology GroupGarvan Institute of Medical ResearchDarlinghurstSydneyNSW2010Australia
- St Vincent's Clinical SchoolFaculty of MedicineUniversity of New South WalesSydneyNSW2010Australia
| | - G. Gregory Neely
- The Dr. John and Anne Chong Laboratory for Functional GenomicsCharles Perkins Centre and School of Life & Environmental SciencesThe University of SydneySydneyNSW2006Australia
| | - Qiao‐Ping Wang
- Laboratory of Metabolism and AgingSchool of Pharmaceutical Sciences (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhen518107China
- Guangdong Provincial Key Laboratory of DiabetologyGuangzhou Key Laboratory of Mechanistic and Translational Obesity ResearchThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
- State Key Laboratory of Anti‐Infective Drug Discovery and DevelopmentSchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| |
Collapse
|
10
|
Kodous AS, Eldin ES, Mohamed HE, Ghobashy MM, El-Maghraby DF. Targeting cell signaling pathway ALKBH5/Beclin1/ULK1 in lung cancer by 5-flurouracil- loaded P (AAm/SA) nanogel in rats. Apoptosis 2025:10.1007/s10495-025-02102-3. [PMID: 40310576 DOI: 10.1007/s10495-025-02102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE Lung cancer is the second most common Cancer in the United States; however, it remains the leading cause of cancer-related death in the United States and worldwide. 5-fluorouracil (5-FU) is among the most administrated chemotherapeutic agents for various neoplasms. This study focused on synthesizing and characterizing P(AAm/SA)/5-Fu nanogels as a potential drug delivery system. METHODS The nanogels were prepared by combining sodium alginate (SA) and acrylamide (AAm) monomers, followed by gamma irradiation-induced polymerization at a dose of 5 kGy. Then, the obtained nanogel was loaded with 500 ppm of 5-Fu. Transmission electron microscopy (TEM) imaging was utilized to characterize the nanogels' morphology and monodispersity with a particle size of (50 nm). Rats were randomly assigned to four groups (six animals per group): Group 1: (Control): normal healthy. Group 2: Cancer-bearing animals (animals injected with diethylnitrosamine (DEN) 20 mg/kg body weight for 3 months. Group 3: Cancer+ 5-fluorouracil (12 mg/kg body weight). Group4: Cancer+ 5-Flurouracil- Loaded P (AAm/SA) Nanogel. RESULTS DEN markedly increased PTGS2, Cox2, PKB, PFKm, and ERK1 levels. Also, observed up-regulation in ALKBH5, Beclin1, ULK1, and P53 gene expressions in the cancer-bearing animal group compared with the control group. 5-fluorouracil nano gel significantly ameliorated the above-mentioned parameters and immunohistochemistry study. 5-fluorouracil nanogel significantly ameliorated the parameters mentioned above, as well as the immunohistochemistry study. CONCLUSION The 5-FU-loaded P(AAm/SA) nanogel could serve as a promising approach for targeting tumor cell proliferation, speeding up autophagic processes, and overcoming chemotherapy resistance in lung carcinoma.
Collapse
Affiliation(s)
- Ahmad S Kodous
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt.
- Department of Molecular Oncology, Cancer Institute (WIA), 38, Sardar Patel Road, P.O. Box 600036, Chennai, Tamilnadu, India.
| | - Eman S Eldin
- Department of Health Radiation Research, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Hebatallah E Mohamed
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Mohamed Mohamady Ghobashy
- Department of of Radiation Research of Polymer Chemistry, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| | - Dina F El-Maghraby
- Department of Health Radiation Research, National Center for Radiation Research and Technology, (NCRRT), Egyptian Atomic Energy Authority (EAEA), P.O. Box 13759, Cairo, Egypt
| |
Collapse
|
11
|
Ding W, Wang L, Wu J, Li H, Li B, Wang Y, Zhang J, Li W. Neuroprotective effects of macrostemonoside T on glutamate-induced injury in HT22 cells. Biochem Pharmacol 2025; 235:116827. [PMID: 39993610 DOI: 10.1016/j.bcp.2025.116827] [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: 10/02/2024] [Revised: 02/12/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Glutamate is a critical excitatory neurotransmitter involved in numerous cellular functions. However, excessive glutamate release can lead to neuronal cell death through oxidative stress, which is implicated in the pathogenesis of various neurological disorders. Therefore, strategies aimed at preventing oxidative stress have emerged as promising therapeutic approaches. Macrostemonoside T (MST), a novel steroidal saponin isolated from the traditional Chinese medicine Allii Macrostemon Bulbus, has demonstrated significant antioxidant activity in previous studies. Nevertheless, its neuroprotective effects against oxidative damage and the underlying molecular mechanisms have not yet been fully elucidated. In this study, we established a glutamate-induced cell injury model using mouse hippocampal neurons (HT22) to investigate the neuroprotective effects of MST and explore its potential mechanisms. A variety of techniques, including DCFH-DA staining, JC-1 staining, Hoechst 33,258 staining, flow cytometry, immunofluorescence staining, ELISA, Western blot analysis, and molecular docking, were employed. The results demonstrated that MST treatment significantly improved the survival of HT22 cells exposed to glutamate. Moreover, MST treatment markedly reduced intracellular levels of reactive oxygen species (ROS) and malondialdehyde while enhancing the activity of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase. MST also mitigated mitochondrial dysfunction by inhibiting mitochondrial fission and preserving mitochondrial membrane potential. Additionally, MST reduced excessive autophagy by decreasing autophagy markers and inhibiting the transition from LC3I to LC3II. Furthermore, MST decreased apoptosis rates, lowered pro-apoptotic protein BAX levels, increased the expression of the anti-apoptotic protein Bcl-2, and inhibited the release of apoptosis-inducing factors from mitochondria. Molecular docking analysis showed that MST enhanced PKA activity by blocking endogenous inhibition of PKA, which in turn activated the PKA/CREB/BDNF signalling pathway. Subsequent validation using immunofluorescence and Western blotting further confirmed that MST treatment significantly reversed the glutamate-induced reduction of PRKACA, CREB, p-CREB, and BDNF protein levels. In conclusion, MST is a potent neuroprotective agent that ameliorates glutamate-induced neuronal damage by inhibiting oxidative stress, alleviating mitochondrial dysfunction, reducing autophagy and apoptosis, and activating the PKA/CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Weixing Ding
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Lulu Wang
- College of Medicine, Changchun Sci-Tech University, Changchun 130600, China
| | - Jianfa Wu
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Hongyan Li
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Bing Li
- College of Medicine, Changchun Sci-Tech University, Changchun 130600, China
| | - Yulei Wang
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jing Zhang
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Wei Li
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, China.
| |
Collapse
|
12
|
Yan W, Xiang S, Feng J, Zu X. Role of ubiquitin-specific proteases in programmed cell death of breast cancer cells. Genes Dis 2025; 12:101341. [PMID: 40083330 PMCID: PMC11904532 DOI: 10.1016/j.gendis.2024.101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 03/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor and the leading cause of cancer-related deaths among women worldwide. Great progress has been recently achieved in controlling breast cancer; however, mortality from breast cancer remains a substantial challenge, and new treatment mechanisms are being actively sought. Programmed cell death (PCD) is associated with the progression and treatment of many types of human cancers. PCD can be divided into multiple pathways including autophagy, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis, and anoikis. Ubiquitination is a post-translational modification process in which ubiquitin, a 76-amino acid protein, is coupled to the lysine residues of other proteins. Ubiquitination is involved in many physiological events and promotes cancer development and progression. This review elaborates the role of ubiquitin-specific protease (USP) in programmed cell death, which is common in breast cancer cells, and lays the foundation for tumor diagnosis and targeted therapy.
Collapse
Affiliation(s)
| | | | - Jianbo Feng
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| | - Xuyu Zu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| |
Collapse
|
13
|
Ivanova T, Sbirkov Y, Kazakova M, Sarafian V. Lysosomes and LAMPs as Autophagy Drivers of Drug Resistance in Colorectal Cancer. Cells 2025; 14:574. [PMID: 40277899 PMCID: PMC12025563 DOI: 10.3390/cells14080574] [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: 02/16/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Colorectal cancer (CRC) is among the most malignant pathologies worldwide. A major factor contributing to the poor prognosis of neoplastic diseases is the development of drug resistance. It significantly reduces the utility of most therapeutic protocols and necessitates the search for novel biomarkers and treatment strategies to combat cancer. An evolutionarily conserved catabolic mechanism, autophagy maintains nutrient recycling and metabolic adaptation and is also closely related to carcinogenesis, playing a dual role. Autophagy inhibition can limit the growth of tumors and improve the response to cancer therapeutics. Lysosomes, key players in autophagy, are also considered promising targets for anticancer treatment. There are still insufficient data on the role of poorly studied glycoproteins related to autophagy, such as the lysosome-associated membrane glycoproteins (LAMPs). They can act as multifunctional molecules involved in a multitude of processes like autophagy and cancer development. In the current review, we summarize the recent data on the double-faceted role of autophagy in cancer with a focus on drug resistance in CRC and on the roles of lysosomes and LAMPs in these interconnected processes. Several lysosomotropic drugs are discussed as options to overcome cancer cell chemoresistance. The complex networks that underline defined autophagic pathways in the context of CRC carcinogenesis and the role of autophagy, especially of LAMPs as drivers of drug resistance, are outlined.
Collapse
Affiliation(s)
- Tsvetomira Ivanova
- Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria; (Y.S.); (M.K.)
- Research Division of Molecular and Regenerative Medicine, Research Institute at Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| | - Yordan Sbirkov
- Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria; (Y.S.); (M.K.)
- Research Division of Molecular and Regenerative Medicine, Research Institute at Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| | - Maria Kazakova
- Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria; (Y.S.); (M.K.)
- Research Division of Molecular and Regenerative Medicine, Research Institute at Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria; (Y.S.); (M.K.)
- Research Division of Molecular and Regenerative Medicine, Research Institute at Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| |
Collapse
|
14
|
Jalali P, Shahmoradi A, Samii A, Mazloomnejad R, Hatamnejad MR, Saeed A, Namdar A, Salehi Z. The role of autophagy in cancer: from molecular mechanism to therapeutic window. Front Immunol 2025; 16:1528230. [PMID: 40248706 PMCID: PMC12003146 DOI: 10.3389/fimmu.2025.1528230] [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: 11/14/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Autophagy is a cellular degradation process that plays a crucial role in maintaining metabolic homeostasis under conditions of stress or nutrient deprivation. This process involves sequestering, breaking down, and recycling intracellular components such as proteins, organelles, and cytoplasmic materials. Autophagy also serves as a mechanism for eliminating pathogens and engulfing apoptotic cells. In the absence of stress, baseline autophagy activity is essential for degrading damaged cellular components and recycling nutrients to maintain cellular vitality. The relationship between autophagy and cancer is well-established; however, the biphasic nature of autophagy, acting as either a tumor growth inhibitor or promoter, has raised concerns regarding the regulation of tumorigenesis without inadvertently activating harmful aspects of autophagy. Consequently, elucidating the mechanisms by which autophagy contributes to cancer pathogenesis and the factors determining its pro- or anti-tumor effects is vital for devising effective therapeutic strategies. Furthermore, precision medicine approaches that tailor interventions to individual patients may enhance the efficacy of autophagy-related cancer treatments. To this end, interventions aimed at modulating the fate of tumor cells by controlling or inducing autophagy substrates necessitate meticulous monitoring of these mediators' functions within the tumor microenvironment to make informed decisions regarding their activation or inactivation. This review provides an updated perspective on the roles of autophagy in cancer, and discusses the potential challenges associated with autophagy-related cancer treatment. The article also highlights currently available strategies and identifies questions that require further investigation in the future.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amir Samii
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Radman Mazloomnejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Afshin Namdar
- Program in Cell Biology, The Hospital for Sick Children Peter Gilgan Centre for Research and Learning, Toronto, ON, United States
| | - Zahra Salehi
- Department of Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Osterli E, Park Y, Hu K, Kasof G, Wiederhold T, Liu C, Hu B. The role of autophagy in ischemic brain injury. AUTOPHAGY REPORTS 2025; 4:2486445. [PMID: 40395988 PMCID: PMC11980474 DOI: 10.1080/27694127.2025.2486445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 05/22/2025]
Abstract
Ischemic brain injury occurs in many clinical settings, including stroke, cardiac arrest, hypovolemic shock, cardiac surgery, cerebral edema, and cerebral vasospasm. Decades of work have revealed many important mechanisms related to ischemic brain injury. However, there remain significant gaps in the scientific knowledge to reconcile many ischemic brain injury events. Brain ischemia leads to protein misfolding and aggregation, and damages almost all types of subcellular organelles including mitochondria, endoplasmic reticulum, Golgi apparatus, lysosomes, etc. Irreparably damaged organelles and insoluble protein aggregates are normally removed by autophagy. The build-up of common autophagic components, such as LC3, p62, and ubiquitinated proteins, are generally observed in brain tissue samples in animal models of both global and focal brain ischemia, but the interpretation of the role of these autophagy-related changes in ischemic brain injury in the literature has been controversial. Many pathological events or mechanisms underlying dysfunctional autophagy after brain ischemia remain unknown. This review aims to provide an update of the current knowledge and future research directions regarding the critical role of dysfunctional autophagy in ischemic brain injury.
Collapse
Affiliation(s)
- Emily Osterli
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Yujung Park
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Kurt Hu
- Department of Medicine, Division of Pulmonary and Critical Care, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Gary Kasof
- Departments of Product Design and Strategy and Antibody Protein Technology, Cell Signaling Technology, Danvers, MA, USA
| | - Thorsten Wiederhold
- Departments of Product Design and Strategy and Antibody Protein Technology, Cell Signaling Technology, Danvers, MA, USA
| | - Chunli Liu
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Bingren Hu
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Research, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| |
Collapse
|
16
|
Dong Y, Gao W, Liu K, Lin C, Usman M, Loor JJ, Li G, Cao L, Yang Z, Luo J, Li D, Sun Q, Lei L, Du X, Song Y, Liu G, Li X. Cullin 3 mitigates nonesterified fatty acid-induced oxidative stress in mammary epithelial cells: Involvement of BCL2/BECN1 and autophagy. J Dairy Sci 2025; 108:4163-4179. [PMID: 40054686 DOI: 10.3168/jds.2024-25879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/04/2025] [Indexed: 04/20/2025]
Abstract
High nonesterified fatty acid (NEFA) concentrations in cows with clinical ketosis lead to metabolic dysfunction in mammary cells, resulting in oxidative stress. Studies have shown that autophagy is impaired in the mammary glands of ketotic cows, and enhancing autophagy mitigates oxidative stress in these animals. Cullin 3 (CUL3), an E3 ubiquitin ligase, is integral for maintaining cellular homeostasis, particularly regulation of oxidative stress and autophagy. Whether CUL3 is involved in mitigating NEFA-induced oxidative stress is unknown. This study aimed to investigate the protective effects and underlying mechanisms whereby CUL3 mitigates NEFA-induced oxidative stress in mammary epithelial cells. First, mammary gland tissue and blood samples were collected from healthy cows (n = 12, BHB <0.6 mM) and cows with clinical ketosis (n = 12, BHB >3.0 mM). Compared with healthy cows, cows with clinical ketosis had reduced productive performance, decreased CUL3 expression, impaired autophagic activity, and increased oxidative stress status in mammary tissue. In vitro, incubating the immortalized bovine mammary epithelial cell line (MAC-T) with 1.2 mM NEFA downregulated CUL3 expression, impaired autophagy, and increased oxidative stress. Adenovirus-mediated overexpression of CUL3 attenuated NEFA-induced accumulation of peroxides and reactive oxygen species, whereas silencing of CUL3 via small interfering RNA exacerbated these effects. Even when nuclear factor erythroid 2 related factor 2 (NFE2L2) expression was reduced by overexpression of CUL3, there was no worsening of NEFA-induced reductions in mRNA levels of NFE2L2 downstream target genes (NADPH quinone oxidoreductase 1 [NQO1], heme oxygenase-1 [HMOX1], glutamate-cysteine ligase catalytic subunit [GCLC)], and glutamate-cysteine ligase modifier subunit [GCLM]). The reduction in NEFA-induced oxidative stress by CUL3 was diminished upon autophagy related 5 (ATG5) silencing suggesting that CUL3 alleviates NEFA-induced oxidative stress via autophagy. Additionally, CUL3 overexpression aggravated the NEFA-induced decrease in BCL2 apoptosis regulator (BCL2) expression along with alleviating the NEFA-induced decrease in Beclin1 (BECN1) expression. Under NEFA treatment, overexpression of BCL2 partly mitigated the CUL3-induced elevation in BECN1. Overall, oxidative stress and impaired autophagy are characterized in the mammary tissue of cows with clinical ketosis. CUL3 activation, likely through the BCL2-BECN1 pathway, enhances autophagy and mitigates NEFA-induced oxidative stress in MAC-T cells. Thus, targeting CUL3-mediated autophagy could be a promising therapeutic strategy to reduce oxidative stress-induced damage in bovine mammary epithelial cells.
Collapse
Affiliation(s)
- Yifei Dong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Kai Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Cai Lin
- College of Food and Biology, Changchun Polytechnic, Changchun City, China 130062
| | - Muhammad Usman
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Juan J Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Guojin Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Liguang Cao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zifeng Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jianchun Luo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Depeng Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Qianwen Sun
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiliang Du
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuxiang Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guowen Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
17
|
Mansour AM, Arafa MM, Hegazy YS, Sadek MS, Ibrahim HH, Abdullah YS, Shehab OR. A comprehensive survey of cytotoxic active half-sandwich Ir(III) complexes: structural perspective, and mechanism of action. Dalton Trans 2025; 54:4788-4847. [PMID: 39932564 DOI: 10.1039/d4dt03219e] [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: 03/18/2025]
Abstract
Iridium(III) complexes, particularly those with piano-stool structures, have drawn a lot of interest recently as possible anticancer drugs. These complexes, which have displayed enhanced cytotoxicity and cytoselectivity compared with clinically approved drugs like cisplatin, oxaliplatin, and carboplatin, hold promising prospects for further anticancer research. Our review aims to explore the complex interplay between cytotoxic properties, cellular uptake efficiency, and intracellular distribution properties of this class of Ir(III) complexes, considering the variation of the coordination site atoms. We provide an overview of the majority of research on mono- and polynunclear half-sandwich Ir(III) complexes with mono- and bidentate ligands, focusing on the impact of altering the leaving group, tethers, substituents on the cyclopentadienyl ring and ligand, spacers, and counter ions on the cytotoxicity and mode of action.
Collapse
Affiliation(s)
- Ahmed M Mansour
- Department of Chemistry, United Arab Emirates University, Al-Ain, United Arab Emirates.
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt
| | - Mohamed M Arafa
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt
| | - Yara S Hegazy
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt
| | - Muhammed S Sadek
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt
| | - Hadeer H Ibrahim
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt
| | - Yomna S Abdullah
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt
| | - Ola R Shehab
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt
| |
Collapse
|
18
|
Li Q, Xiao N, Zhang H, Liang G, Lin Y, Qian Z, Yang X, Yang J, Fu Y, Zhang C, Liu A. Systemic aging and aging-related diseases. FASEB J 2025; 39:e70430. [PMID: 40022602 DOI: 10.1096/fj.202402479rrr] [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: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Aging is a biological process along with systemic and multiple organ dysfunction. It is more and more recognized that aging is a systemic disease instead of a single-organ functional disorder. Systemic aging plays a profound role in multiple diseases including neurodegenerative diseases, cardiovascular diseases, and malignant diseases. Aged organs communicate with other organs and accelerate aging. Skeletal muscle, heart, bone marrow, skin, and liver communicate with each other through organ-organ crosstalk. The crosstalk can be mediated by metabolites including lipids, glucose, short-chain fatty acids (SCFA), inflammatory cytokines, and exosomes. Metabolic disorders including hyperglycemia, hyperinsulinemia, and hypercholesterolemia caused by chronic diseases accelerate hallmarks of aging. Systemic aging leads to the destruction of systemic hemostasis, causes the release of inflammatory cytokines, senescence-associated secretory phenotype (SASP), and the imbalance of microbiota composition. Released inflammatory factors further aggregate senescence, which promotes the aging of multiple solid organs. Targeting senescence or delaying aging is emerging as a critical health strategy for solving age-related diseases, especially in the old population. In the current review, we will delineate the mechanisms of organ crosstalk in systemic aging and age-related diseases to provide therapeutic targets for delaying aging.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yanguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
19
|
Siedlecka-Kroplewska K, Kmiec Z, Zmijewski MA. The Interplay Between Autophagy and Apoptosis in the Mechanisms of Action of Stilbenes in Cancer Cells. Antioxidants (Basel) 2025; 14:339. [PMID: 40227400 PMCID: PMC11939748 DOI: 10.3390/antiox14030339] [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: 01/31/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Plant-based stilbenes are low-molecular-weight polyphenolic compounds that exhibit anti-oxidant, anti-microbial, anti-fungal, anti-inflammatory, anti-diabetic, cardioprotective, neuroprotective, and anti-cancer activities. They are phytoalexins produced in diverse plant species in response to stress, such as fungal and bacterial infections or excessive UV irradiation. Plant-derived dietary products containing stilbenes are common components of the human diet. Stilbenes appear to be promising chemopreventive and chemotherapeutic agents. Accumulating evidence indicates that stilbenes are able to trigger both apoptotic and autophagic molecular pathways in many human cancer cell lines. Of note, the molecular crosstalk between autophagy and apoptosis under cellular stress conditions determines the cell fate. The autophagy and apoptosis relationship is complex and depends on the cellular context, e.g., cell type and cellular stress level. Apoptosis is a type of regulated cell death, whereas autophagy may act as a pro-survival or pro-death mechanism depending on the context. The interplay between autophagy and apoptosis may have an important impact on chemotherapy efficiency. This review focuses on the in vitro effects of stilbenes in different human cancer cell lines concerning the interplay between autophagy and apoptosis.
Collapse
Affiliation(s)
| | - Zbigniew Kmiec
- Department of Anatomy and Histology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland;
| | | |
Collapse
|
20
|
Bustamante M, Quiroga C, Mancilla G, Gomez W, Tapia A, Figueroa R, Mondaca-Ruff D, Oyarzún I, Verdejo HE, Lavandero S, Castro P. Autophagy fine-tuning by angiotensin-(1-9) in cultured rat cardiomyocytes. Front Cardiovasc Med 2025; 12:1408325. [PMID: 40144934 PMCID: PMC11937029 DOI: 10.3389/fcvm.2025.1408325] [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: 03/28/2024] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Background The renin-angiotensin system (RAS) plays a pivotal role in regulating blood volume, systemic vascular resistance, and electrolyte balance, serving as a key component of cardiovascular health. Recent findings highlight the role of angiotensin II (Ang II) in inducing autophagy through angiotensin II receptor type 1 (AT1R). Autophagy, a process of self-degradation and turnover of cellular components, is a homeostatic response that eliminates superfluous materials. Abnormal autophagy promotes cardiomyocyte loss and is critical in hypertrophy and heart failure progression. The RAS's non-canonical axis, which includes the angiotensin 1-9 peptide [Ang-(1-9)], has an anti-hypertrophic effect in cardiomyocytes via an unknown mechanism. In the present study, we aimed to elucidate the effect of Ang-(1-9) on cardiomyocyte autophagy. Methods We isolated and cultured neonatal ventricular cardiomyocytes and then co-treated them with Ang-(1-9) in the presence of chloroquine (CQ), Ang-II, and chemical inhibitors of different signaling pathways. After treatment, total RNA and protein extracts were obtained to analyze the abundance of different autophagy markers. Likewise, cells were fixed, and autophagy was analyzed through epifluorescence microscopy. Results Our findings show that CQ leads to a reduction in autophagy markers, such as microtubule-associated protein 1 light chain 3-II (LC3-II) and total LC3, suggesting Ang-(1-9)'s regulatory role in basal autophagy levels. Furthermore, Ang-(1-9) opposes Ang-II-induced autophagy and induces the phosphorylation of the S234 residue of Beclin-1 (BCN1) via an angiotensin II receptor type 2 (AT2R)/Akt-dependent pathway. Conclusions This reduction of Ang-II-induced autophagy by Ang-(1-9) unveils a novel aspect of its action, potentially contributing to its cardioprotective effects.
Collapse
Affiliation(s)
- Mario Bustamante
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Clara Quiroga
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Georthan Mancilla
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Physiology and Biophysics Program, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Wileidy Gomez
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Anita Tapia
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Reinaldo Figueroa
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David Mondaca-Ruff
- Department of Biochemistry and Molecular Biology & Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ingrid Oyarzún
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo E. Verdejo
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Transducción de Señales Moleculares, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Department of Internal Medicine/Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Pablo Castro
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile & Pontifical Catholic University of Chile, Santiago, Chile
- Laboratorio de Señalización Cardiovascular, División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
21
|
Stott-Marshall RJ, McBeth C, Wileman T. Dynamic regulation of autophagy during Semliki Forest virus infection of neuroblastoma cells. J Gen Virol 2025; 106:002086. [PMID: 40042894 PMCID: PMC11882037 DOI: 10.1099/jgv.0.002086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/19/2025] [Indexed: 05/13/2025] Open
Abstract
Autophagy can defend against infection by delivering viruses to lysosomes for degradation. Semliki Forest virus (SFV) is a positive-sense, single-stranded RNA virus of the alphavirus genus which has been used extensively as a model for arbovirus infection and neuronal encephalitis. Here, we show that autophagy is suppressed during the early hours of SFV infection of neurons. We also show that a switch between autophagy suppression and upregulation between the early and later stages was mediated through modulation of the mammalian target of rapamycin (mTOR) activity during infection. At later stages of infection, autophagosomes colocalize with SFV nonstructural proteins suggesting the formation of a platform for virus replication. Inhibition of mTOR by torin reduced infectious virus production and intracellular virus gene expression while improving cell survival during infection. The results suggest that autophagy is suppressed early during infection of neurons to increase cell survival and then upregulated at later times to facilitate replication. This biphasic regulation of autophagy seen for SFV may be important for other arboviruses, and knowledge about the regulation of autophagy by alphaviruses may be useful for the development of antiviral therapies.
Collapse
Affiliation(s)
- Robert J. Stott-Marshall
- One Virology, School of Veterinary Science and Medicine, University of Nottingham, Nottingham, LE12 5RD, UK
- School of Health, Leeds Beckett University, Leeds, LS1 3HE, UK
| | - Craig McBeth
- School of Health and Life Sciences, Teesside University, Middlesbrough, TS1 3BX, UK
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, UK
| |
Collapse
|
22
|
Pal C. Mitochondria-targeting by small molecules against Alzheimer's disease: A mechanistic perspective. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167617. [PMID: 39647244 DOI: 10.1016/j.bbadis.2024.167617] [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: 08/06/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
Alzheimer's disease (AD) poses a considerable worldwide health obstacle, marked by gradual cognitive deterioration and neuronal loss. While the molecular mechanisms underlying AD pathology have been elucidated to some extent, therapeutic options remain limited. Mitochondrial dysfunction has become recognized as a significant factor in the development of AD, with oxidative stress and disrupted energy metabolism being critical elements. This review explores the mechanistic aspects of small molecule targeting of mitochondria as a potential therapeutic approach for AD. The review explores the role of mitochondrial dysfunction in AD, including its involvement in the accumulation of β-amyloid plaques and neurofibrillary tangles, synaptic dysfunction, and neuronal death. Furthermore, the effects of oxidative stress on mitochondrial function were investigated, including the resulting damage to mitochondrial components. Mitochondrial-targeted therapies have attracted attention for their potential to restore mitochondrial function and reduce AD pathology. The review outlines the latest preclinical and clinical evidence supporting the effectiveness of small molecules in targeting mitochondrial dysfunction in AD. Additionally, it discusses the molecular pathways involved in mitochondrial dysfunction and examines how small molecules can intervene to address these abnormalities. By providing a comprehensive overview of the latest research in this field, this review aims to shed light on the therapeutic potential of small molecule targeting of mitochondria in AD and stimulate further research in this promising area of drug development.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Chemistry, Gobardanga Hindu College, North 24 Parganas, West Bengal 743273, India.
| |
Collapse
|
23
|
Vona R, Cittadini C, Ortona E, Matarrese P. Sex Disparity in Cancer: Role of Autophagy and Estrogen Receptors. Cells 2025; 14:273. [PMID: 39996745 PMCID: PMC11854201 DOI: 10.3390/cells14040273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Autophagy, a cellular process essential for maintaining homeostasis, plays a fundamental role in recycling damaged components and in adapting to stress. The dysregulation of autophagy is implicated in numerous human diseases, including cancer, where it exhibits a dual role as both a suppressor and a promoter, depending on the context and the stage of tumor development. The significant sex differences observed in autophagic processes are determined by biological factors, such as genetic makeup and sex hormones. Estrogens, through their interaction with specific receptors, modulate autophagy and influence tumor progression, therapy resistance, and the immune response to tumors. In females, the escape from X inactivation and estrogen signaling may be responsible for the advantages, in terms of lower incidence and longer survival, observed in oncology. Women often show better responses to traditional chemotherapy, while men respond better to immunotherapy. The action of sex hormones on the immune system could contribute to these differences. However, women experience more severe adverse reactions to anticancer drugs. The estrogen/autophagy crosstalk-involved in multiple aspects of the tumor, i.e., development, progression and the response to therapy-deserves an in-depth study, as it could highlight sex-specific mechanisms useful for designing innovative and gender-tailored treatments from the perspective of precision medicine.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| | | | | | - Paola Matarrese
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| |
Collapse
|
24
|
Saluja TS, Hosalkar R. Prognostic Utility of Autophagy Marker Beclin1 in Oral Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis. Head Neck Pathol 2025; 19:17. [PMID: 39907919 PMCID: PMC11799460 DOI: 10.1007/s12105-025-01755-5] [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: 11/16/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Autophagy is involved in several critical cellular processes regulating cell survival and death. Past research suggests that it may either act as a tumor suppressor or promote tumor progression. The purpose of this systematic review and meta-analysis was to evaluate the clinical and prognostic utility of a significant autophagy related protein-Beclin1, in oral squamous cell carcinoma (OSCC). METHODS Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines were followed. Relevant literature was retrieved from PubMed, ScienceDirect and Google Scholar database. After removal of duplicates quality of the studies was assessed using Newcastle-Ottawa Scale. Heterogeneity was assessed using I2 index. Random effect model was used if I2 was more than 50% else fixed effect model was selected. Meta-analysis was carried out using Review Manager (RevMan; Version 5.4). RESULTS Five studies with 494 cases were included in this meta-analysis. Beclin1 expression in OSCC was not significantly associated (p > 0.05) with gender, age, tumor size, lymph node metastasis, histological differentiation and overall survival. Nevertheless, a trend for low Beclin1 expression favoring tumor progression was observed. Sensitivity analysis revealed significant nodal positivity related to low Beclin1 expression. CONCLUSION This study provided an overview of Beclin1 expression in OSCC and highlighted additional evaluations while its use as a prognostic marker. It is suggested that future studies should assess both nuclear as well as cytoplasmic expression of Beclin1 and report intra- and inter-tumor variations in its expression relating to clinicopathological parameters.
Collapse
Affiliation(s)
- Tajindra Singh Saluja
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Rashmi Hosalkar
- Department of Oral Pathology and Microbiology, MGM Dental College and Hospital, MGM Institute of Health Sciences, Navi Mumbai, Maharashtra, India.
| |
Collapse
|
25
|
Kausar MA, Anwar S, Khan YS, Saleh AA, Ahmed MAA, Kaur S, Iqbal N, Siddiqui WA, Najm MZ. Autophagy and Cancer: Insights into Molecular Mechanisms and Therapeutic Approaches for Chronic Myeloid Leukemia. Biomolecules 2025; 15:215. [PMID: 40001518 PMCID: PMC11853340 DOI: 10.3390/biom15020215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Autophagy is a critical cellular process that maintains homeostasis by recycling damaged or aberrant components. This process is orchestrated by a network of proteins that form autophagosomes, which engulf and degrade intracellular material. In cancer, autophagy plays a dual role: it suppresses tumor initiation in the early stages but supports tumor growth and survival in advanced stages. Chronic myeloid leukemia (CML), a hematological malignancy, is characterized by the Philadelphia chromosome, a chromosomal abnormality resulting from a translocation between chromosomes 9 and 22. Autophagy has emerged as a key factor in CML pathogenesis, promoting cancer cell survival and contributing to resistance against tyrosine kinase inhibitors (TKIs), the primary treatment for CML. Targeting autophagic pathways is being actively explored as a therapeutic approach to overcome drug resistance and enhance cancer cell death. Recent research highlights the intricate interplay between autophagy and CML progression, underscoring its role in disease biology and treatment outcomes. This review aims to provide a comprehensive analysis of the molecular and cellular mechanisms underlying CML, with a focus on the therapeutic potential of targeting autophagy.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Autophagy/drug effects
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Animals
- Drug Resistance, Neoplasm/drug effects
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Yusuf Saleem Khan
- Department of Anatomy, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | - Ayman A. Saleh
- Department of Pathology, College of Medicine, University of Ha’il, Hail 55476, Saudi Arabia;
| | | | - Simran Kaur
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122103, Haryana, India;
| | - Naveed Iqbal
- Department of Obstetrics and Gynecology, College of Medicine, University of Ha’il, Ha’il 55476, Saudi Arabia;
| | - Waseem Ahmad Siddiqui
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202001, Uttar Pradesh, India;
| | - Mohammad Zeeshan Najm
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122103, Haryana, India;
| |
Collapse
|
26
|
Qian S, Tan G, Lei G, Zhang X, Xie Z. Programmed cell death in nasopharyngeal carcinoma: Mechanisms and therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189265. [PMID: 39809344 DOI: 10.1016/j.bbcan.2025.189265] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Programmed cell death is a type of autonomic and orderly cell death mode controlled by genes that maintain homeostasis and growth. Tumor is a typical manifestation of an imbalance in environmental homeostasis in the human body. Currently, several tumor treatments are designed to trigger the death of tumor cells. Nasopharyngeal carcinoma is one of the most common malignant tumors in China. It displays obvious regional and ethnic differences in its incidence, being typically high in the south and low in the north of China. Nasopharyngeal carcinoma is currently considered to be a polygenic inherited disease and is often mediated by the interaction between multiple genes or between genes and the environment. Apoptosis has long been considered the key to tumor treatment, while other cell death pathways have often been overlooked. The current study provides an overview of the relationship among apoptosis, autophagy, pyroptosis, necroptosis, ferroptosis, and nasopharyngeal carcinoma, and the regulatory pathways of nasopharyngeal carcinoma based on five cell death modes were synthesized from the view of molecule. We hope this review will help explore additional, novel programmed cell death targets for the treatment of nasopharyngeal carcinoma and thus promote in-depth research.
Collapse
Affiliation(s)
- Shen'er Qian
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guolin Tan
- Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guang Lei
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School Of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaowei Zhang
- Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zuozhong Xie
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Laboratory of Otolaryngology Head and Neck Cancer, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Otology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Chen Y, Yi H, Liao S, He J, Zhou Y, Lei Y. LC3B: A microtubule-associated protein influences disease progression and prognosis. Cytokine Growth Factor Rev 2025; 81:16-26. [PMID: 39701849 DOI: 10.1016/j.cytogfr.2024.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024]
Abstract
Microtubule-associated protein 1 light chain 3B (MAP1LC3B, also known as LC3B) is a mammalian homolog of the autophagy-related protein 8 (ATG8) family. It plays a crucial role in cellular autophagy and is involved in several vital biological processes, including apoptosis and differentiation. Additionally, LC3B regulates immune responses. Due to its close association with malignant tumors and neurodegenerative diseases, and its potential as a prognostic indicator and therapeutic target, LC3B has become a significant research focus. This article aims to provide a comprehensive and systematic understanding of LC3B's role and mechanisms in autophagy, its impact on apoptosis and the underlying mechanisms, its regulation of cellular differentiation and transdifferentiation, its modulation of immune and inflammatory responses, the influence of upstream regulatory factors on LC3B's function, and its relevance to disease diagnosis, treatment, and prognosis. The goal is to establish a solid foundation for understanding LC3B's role in cellular processes and its regulatory mechanisms.
Collapse
Affiliation(s)
- Yan Chen
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Hong Yi
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Shan Liao
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Junyu He
- Department of Clinical Laboratory, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan 410007, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China.
| | - Yan Lei
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, Hunan 410013, China.
| |
Collapse
|
28
|
Zhao H, Yang M, Han Y, Jiang N, Liu Y, Li C, Yang J, Luo S, Liu C, Sun L, Liu F, Liu Y. HIF-1α/BNIP3-Mediated Endoplasmic Reticulum Degradation via Autophagy Protects Against Ischemia Reperfusion-Induced Acute Kidney Injury. Antioxid Redox Signal 2025; 42:212-227. [PMID: 39099334 DOI: 10.1089/ars.2023.0467] [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] [Indexed: 08/06/2024]
Abstract
Aims: Endoplasmic reticulum (ER) degradation via autophagy is a process that maintains ER homeostasis when cells are in a state of stress and is associated with many diseases; however, the role of hypoxia inducible factor-1α (HIF-1α)-mediated ER degradation and the related regulatory pathway in acute kidney injury (AKI) still needs to be further established. Results: In the present study, an in vivo AKI model was induced in mice via the ischemia-reperfusion (IR) method. The results revealed that HIF-1α and BNIP3 were increased, and autophagy and ER degradation were activated in the kidneys of AKI mice, whereas HIF-1α knockout significantly inhibited BNIP3, autophagy and ER degradation, accompanied by aggravated kidney injury. Overexpression of HIF-1α in vitro significantly increased BNIP3, autophagy and ER degradation, whereas inhibition of BNIP3 significantly reversed the effects of HIF-1α. In addition, the in vitro inhibition of autophagy with chloroquine significantly reversed the effects of HIF-1α on cell apoptosis. Moreover, selectively overexpressing BNIP3 on the ER membrane significantly increased ER degradation via autophagy and decreased cell apoptosis in vitro. Innovation and Conclusion: These data indicate that HIF-1α/BNIP3-mediated ER degradation via autophagy in tubular cells protects against IR-induced AKI. Antioxid. Redox Signal. 42, 212-227.
Collapse
Affiliation(s)
- Hao Zhao
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Ming Yang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Na Jiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chenrui Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinfei Yang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shilu Luo
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chongbin Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
29
|
Güler A, Yardımcı BK, Özek NŞ. Human anti-apoptotic Bcl-2 and Bcl-xL proteins protect yeast cells from aging induced oxidative stress. Biochimie 2025; 229:69-83. [PMID: 39413900 DOI: 10.1016/j.biochi.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/24/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Aging is a degenerative, biological, and time-dependent process that affects all organisms. Yeast aging is a physiological phenomenon characterized by the progressive transformation of yeast cells, resulting in modifications to their viability and vitality. Aging in yeast cells is comparable to that in higher organisms in some respects; however, due to their straightforward and well-characterized genetic makeup, these cells present unique advantages when it comes to researching the aging process. Here, we assessed the impact of human anti-apoptotic Bcl-2 and Bcl-xL proteins on aging using a yeast model. The findings clearly showed that these proteins exhibited remarkable anti-aging properties in yeast cells. Our data indicate that the presence of both proteins enhanced the reproductive survival of aging cells, likely by effecting the components functioning as both pro- and anti-oxidants, depending on the stage of yeast cell lifespan. Both proteins partially protected yeast cells from aging-related morphological deformations and cellular damage during the aging period. In particular, Bcl-xL expressing yeast cells reached the maximum activity levels for almost all of the major antioxidant enzymes and the total antioxidant status on the 8th day of lifespan and could provide effective protection at the latest stage of the investigated aging period. The chemometric data analysis of IR spectra confirmed the findings of the morphological and biochemical analyses. In this regard, specifically, understanding the mechanism of action on the cellular redox state of Bcl-xL in yeast may facilitate comprehension of its indirect antioxidant function in higher eukaryotes.
Collapse
Affiliation(s)
- Ayşenur Güler
- Chemistry Department, Graduate School of Natural and Applied Sciences, Pamukkale University, Denizli, Turkey
| | - Berna Kavakcıoğlu Yardımcı
- Department of Chemistry, Faculty of Science, Pamukkale University, Denizli, Turkey; Advanced Technology Application and Research Center, Pamukkale University, Denizli, Turkey.
| | - Nihal Şimşek Özek
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey; East Anatolian High Technology Research and Application Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
30
|
Sabry NC, Michel HE, Menze ET. Repurposing of erythropoietin as a neuroprotective agent against methotrexate-induced neurotoxicity in rats. J Psychopharmacol 2025; 39:147-163. [PMID: 39535118 DOI: 10.1177/02698811241295379] [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] [Indexed: 11/16/2024]
Abstract
BACKGROUND Methotrexate (MTX) is a cytotoxic drug that can trigger neurotoxicity via enhancing oxidative stress, apoptosis, and inflammation. On the other hand, erythropoietin (EPO) functions as an antioxidant, anti-apoptotic, and anti-inflammatory agent, in addition to its hematopoietic effects. AIM The present study was developed to examine the neuroprotective impact of EPO against MTX-provoked neurotoxicity in rats. METHODS Chemo fog was elicited in Wistar rats via injection of one dosage of MTX (20 mg/kg, i.p) on the sixth day of the study. EPO was injected at 500 IU/kg/day, i.p for 10 successive days. RESULTS MTX triggered memory and learning impairment as evidenced by Morris water maze, passive avoidance, and Y-maze cognitive tests. In addition, MTX induced oxidative stress as evident from the decline in hippocampal Nrf2 and HO-1 levels. MTX brought about apoptosis, as demonstrated by the elevation in p53, caspase-3, and Bax levels, as well as the decrease in Bcl2 levels. MTX also decreased Beclin-1, an autophagy-related marker, and increased P62 expression. In addition, MTX downregulated Sirt-1/AKT/FoxO3a pathway and increased miRNA-34a gene expression. Moreover, MTX increased acetylcholinesterase activity and reduced neurogenesis. EPO administration remarkably counteracted MTX-induced molecular and behavioral disorders in rat hippocampi. CONCLUSION Our findings impart preclinical indication for repurposing of EPO as a promising neuroprotective agent through modulating miRNA-34a, autophagy, and the Sirt-1/FoxO3a signaling pathway.
Collapse
Affiliation(s)
- Nadine C Sabry
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Ain Shams University, Cairo, Egypt
| | - Esther T Menze
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Ain Shams University, Cairo, Egypt
| |
Collapse
|
31
|
Verçosa BLA, Muniz-Junqueira MI, Mineiro ALBB, Melo MN, Vasconcelos AC. Enhanced apoptosis and inflammation allied with autophagic and apoptotic Leishmania amastigotes in the seemingly undamaged ear skin of clinically affected dogs with canine visceral Leishmaniasis. Cell Immunol 2025; 408:104909. [PMID: 39701006 DOI: 10.1016/j.cellimm.2024.104909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Programmed cell death plays a relevant role in the pathogenesis of visceral Leishmaniasis. Apoptosis selects suitable parasites, regulating parasite density, whereas autophagy eliminates pathogens. This study aimed to assess the inflammation and apoptosis in inflammatory cells and presents a unique description of the presence of autophagic and apoptotic Leishmania amastigotes in naturally Leishmania-infected dogs. Fragments from seemingly undamaged ear skin of sixteen Leishmania-infected dogs and seven uninfected dogs were evaluated through histomorphometry, ultrastructural, immunohistochemical and transmission electron microscopy (TEM) analyses. Leishmania amastigotes were present on seemingly undamaged ear skin only in clinically affected dogs. Parasite load, morphometrical parameters of inflammation and apoptotic index of inflammatory cells were higher in clinically affected animals and were related to clinical manifestations. Apoptotic index and morphometric parameters of the inflammatory infiltrate in undamaged ear skin were positively correlated with parasite load. Apoptotic and non-apoptotic Leishmania amastigotes were observed within neutrophils and macrophages. Leishmania amastigotes were positive for Bax, a marker for apoptosis, by immunohistochemistry. Morphological characteristics of apoptosis and autophagy in Leishmania amastigotes were observed only in phagocytes of clinically affected dogs. Positive correlations were found between histomorphometry and clinical manifestations. Our results showed that apoptosis and autophagy in Leishmania amastigotes may be related to both the increase in parasite load and apoptotic index in inflammatory cells, and with the intensity of the inflammatory response in clinically affected dogs. Thus, our study suggests that apoptotic and autophagy Leishmania within phagocytes may have facilitate the survival of the parasite and it appears to play an important role in the process of Leishmania infection.
Collapse
Affiliation(s)
- Barbara Laurice Araújo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil; Faculdade de Ciências da Saúde Pitágoras, Campus Codó, Codó, Maranhão, Brazil.
| | | | - Ana Lys Bezerra Barradas Mineiro
- Departamento de Clínica e Cirurgia Veterinária, Centro de Ciências Agrárias, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
32
|
Wang L, Shi R, Wang S, Duan Y, Wang Z, Zheng P, Sun X, Chen X, Ji G, Shen Y, Dong B, Lin Y, Wen T, Tian Q, Guo Z, Hou Y, Wu S, Xiao L, Li M, Xiao L, Wu Q, Meng Y, Liu G, Duan S, Bai X, Liu T, Zhang Z, Zhan P, Lu Z, Xu D. ADSL promotes autophagy and tumor growth through fumarate-mediated Beclin1 dimethylation. Nat Chem Biol 2025:10.1038/s41589-024-01825-9. [PMID: 39881212 DOI: 10.1038/s41589-024-01825-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025]
Abstract
As an enzyme with a critical role in de novo purine synthesis, adenylosuccinate lyase (ADSL) expression is upregulated in various malignancies. However, whether ADSL possesses noncanonical functions that contribute to cancer progression remains poorly understood. Here, we demonstrate that protein kinase R-like endoplasmic reticulum kinase (PERK) activated by lipid deprivation or ER stress phosphorylates ADSL at S140, leading to an enhanced association between ADSL and Beclin1. Beclin1-associated ADSL produces fumarate, which in turn inhibits lysine demethylase 8-mediated Beclin1 demethylation, resulting in enhanced Beclin1 K117me2, subsequent disruption of the binding of BCL-2 to Beclin1 and elevated autophagy. Blocking the ADSL-Beclin1 axis by knock-in mutation or a cell-penetrating peptide inhibits autophagy induced by lipid deprivation and ER stress and blunts liver tumor growth in mice. Additionally, ADSL pS140-upregulated Beclin1 K117me2 levels are positively correlated with autophagy levels in human hepatocellular carcinoma specimens and poor patient prognosis. These findings uncover the function of ADSL in autophagy regulation and liver tumor development.
Collapse
Affiliation(s)
- Lei Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Runze Shi
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yuran Duan
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Peixiang Zheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guimei Ji
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Yuli Shen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Bofei Dong
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Yanni Lin
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Ting Wen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Qi Tian
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Zhanpeng Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Yueru Hou
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Shiqi Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Ling Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Min Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Qingang Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Guijun Liu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
| | - Sofie Duan
- Canyon Crest Academy, San Diego, CA, USA
| | - Xueli Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Tong Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
33
|
Yu Z, Fan C, Mao Y, Wu X, Mao H. Autophagy activation alleviates annulus fibrosus degeneration via the miR-2355-5p/mTOR pathway. J Orthop Surg Res 2025; 20:86. [PMID: 39849546 PMCID: PMC11755947 DOI: 10.1186/s13018-025-05492-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/12/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Intervertebral disc degeneration disease (IVDD) is a major cause of disability and reduced work productivity worldwide. Annulus fibrosus degeneration is a key contributor to IVDD, yet its mechanisms remain poorly understood. Autophagy, a vital process for cellular homeostasis, involves the lysosomal degradation of cytoplasmic proteins and organelles. This study aimed to investigate the role of autophagy in IVDD using a hydrogen peroxide (H2O2)-induced model of rat annulus fibrosus cells (AFCs). METHODS AFCs were exposed to H2O2 to model oxidative stress-induced degeneration. Protein expression levels of collagen I, collagen II, MMP3, and MMP13 were quantified. GEO database analysis identified alterations in miR-2355-5p expression, and its regulatory role on the mTOR pathway and autophagy was assessed. Statistical tests were used to evaluate changes in protein expression and pathway activation. RESULTS H2O2 exposure reduced collagen I and collagen II expression to approximately 50% of baseline levels, while MMP3 and MMP13 expression increased twofold. Activation of autophagy restored collagen I and II expression and decreased MMP3 and MMP13 levels. GEO analysis revealed significant alterations in miR-2355-5p expression, confirming its role in regulating the mTOR pathway and autophagy. CONCLUSIONS Autophagy, mediated by the miR-2355-5p/mTOR pathway, plays a protective role in AFCs degeneration. These findings suggest a potential therapeutic target for mitigating IVDD progression.
Collapse
Affiliation(s)
- Zilin Yu
- Orthopedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214026, Jiangsu, China
| | - Chunyang Fan
- Orthopedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yubo Mao
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214026, Jiangsu, China.
| | - Xiexing Wu
- Orthopedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Haiqing Mao
- Orthopedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
34
|
Chan WWR, Chow J, Chau DDL, Zhai Y, Lau KF. Beclin 1-Mediated Autophagy Is Potentiated by an Interaction with the Neuronal Adaptor FE65. BIOLOGY 2025; 14:97. [PMID: 39857327 PMCID: PMC11763304 DOI: 10.3390/biology14010097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Autophagy is a vital cellular pathway in eukaryotic cells, including neurons, where it plays significant roles in neurodevelopment and maintenance. A crucial step in autophagy is the formation of the class III phosphatidylinositol 3-kinase complex 1 (PI3KC3-C1), which is essential for initiating autophagosome biogenesis. Beclin 1 is the key component of PI3KC3-C1, and its interactors have been reported to affect autophagy. The brain-enriched adaptor protein FE65 has been shown to interact with Alzheimer's disease amyloid precursor protein (APP) to alter the processing of APP. Additionally, FE65 has been implicated in various cellular pathways, including autophagy. We demonstrate here that FE65 positively regulates autophagy. FE65, through its C-terminus, has been shown to interact with Beclin 1. Notably, the overexpression of FE65 enhances Beclin 1-mediated autophagy, whereas this process is attenuated in FE65 knockout cells. Moreover, the stimulatory effect of FE65 on Beclin 1-mediated autophagy is diminished by an FE65 C-terminus deletion mutant that disrupts the FE65-Beclin 1 interaction. Lastly, we have found that the FE65-Beclin 1 interaction modulates the kinase activity of the PI3KC3-C1 complex. Together, we have identified FE65 as a novel Beclin 1 interactor, and this interaction potentiates autophagy.
Collapse
Affiliation(s)
| | | | | | | | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China; (W.W.R.C.); (J.C.); (D.D.-L.C.); (Y.Z.)
| |
Collapse
|
35
|
Perez-Serna AA, Guzman-Llorens D, Dos Santos RS, Marroqui L. Bcl-2 and Bcl-xL in Diabetes: Contributions to Endocrine Pancreas Viability and Function. Biomedicines 2025; 13:223. [PMID: 39857806 PMCID: PMC11760435 DOI: 10.3390/biomedicines13010223] [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: 12/13/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes is a chronic metabolic disorder whose prevalence increases every year, affecting more than 530 million adults worldwide. Type 1 (T1D) and type 2 diabetes (T2D), the most common forms of diabetes, are characterized by the loss of functional pancreatic β-cells, mostly due to apoptosis. B-cell leukemia/lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xL), two anti-apoptotic proteins belonging to the Bcl-2 family, are crucial for regulating the intrinsic pathway of apoptosis. However, over the years, they have been implicated in many other cellular processes, including intracellular Ca2+ homeostasis and the regulation of mitochondrial metabolism. Thus, understanding the biological processes in which these proteins are involved may be crucial to designing new therapeutic targets. This review summarizes the roles of Bcl-2 and Bcl-xL in apoptosis and metabolic homeostasis. It focuses on how the dysregulation of Bcl-2 and Bcl-xL affects pancreatic β-cell function and survival, and the consequences for diabetes development.
Collapse
Affiliation(s)
- Atenea A. Perez-Serna
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Guzman-Llorens
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
| | - Reinaldo S. Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l’Almazara 11, 03203 Elche, Alicante, Spain
| | - Laura Marroqui
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
36
|
Selim SM, El Fayoumi HM, El-Sayed NM, Mehanna ET, Hazem RM. Alogliptin attenuates STZ-induced diabetic nephropathy in rats through the modulation of autophagy, apoptosis, and inflammation pathways: Targeting NF-κB and AMPK/mTOR pathway. Life Sci 2025; 361:123307. [PMID: 39662777 DOI: 10.1016/j.lfs.2024.123307] [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: 09/24/2024] [Revised: 11/24/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
AIM Diabetic nephropathy (DN) is a type of microvascular complication that arises from diabetes mellitus and leads to further health issues. Most importantly, the prevalence of DN is steadily rising in developed countries. This research explored the therapeutic benefits of alogliptin, a dipeptidyl peptidase IV (DPP-4) inhibitor, on streptozotocin (STZ)-induced DN and its underlying mechanisms in rats. MAIN METHODS Ten rats were allocated to group 1, served as the normal group; and received saline. To develop diabetes, thirty rats were administered a single intraperitoneal dose of STZ (45 mg/kg). STZ-induced diabetic rats were randomly assigned to three groups: group 2 diabetic control; was given saline, groups 3 and 4 received alogliptin (10 mg/kg) and (20 mg/kg), respectively. The treatment began 8 weeks after diabetes onset and continued for four weeks. Histopathological alterations in the kidney were detected. Serum was collected to measure blood glucose levels (BGL), renal function, and lactate dehydrogenase (LDH). Tissue samples were collected to detect changes in oxidative stress (OS), inflammation, 5' adenosine monophosphate-activated protein kinase (AMPK), and the mammalian target of Rapamycin (mTOR) signaling pathways in addition to apoptotic and autophagy changes. KEY FINDINGS Alogliptin reduced STZ-induced histological changes in the kidney as well as OS, and inflammation. Alogliptin also ameliorated the AMPK/mTOR signaling pathways, enhanced autophagy, and reduced apoptosis. SIGNIFICANCE These results demonstrate that alogliptin ameliorates inflammation and OS and consequently modulates the AMPK/mTOR axis along with targeting autophagy and apoptosis, leading to the alleviation of DN.
Collapse
Affiliation(s)
- Salma M Selim
- Department of Pharmacology & Toxicology, Faculty of Dentistry, Sinai University, Kantara, Ismailia 41636, Egypt
| | - Hassan M El Fayoumi
- Department of Pharmacology & Toxicology, Faculty of Dentistry, Sinai University, Kantara, Ismailia 41636, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| | - Reem M Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
37
|
Taha SR, Karimi M, Mahdavi B, Yousefi Tehrani M, Bemani A, Kabirian S, Mohammadi J, Jabbari S, Hushmand M, Mokhtar A, Pourhanifeh MH. Crosstalk between non-coding RNAs and programmed cell death in colorectal cancer: implications for targeted therapy. Epigenetics Chromatin 2025; 18:3. [PMID: 39810224 PMCID: PMC11734566 DOI: 10.1186/s13072-024-00560-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most common causes of cancer-related mortality worldwide. Its progression is influenced by complex interactions involving genetic, epigenetic, and environmental factors. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been identified as key regulators of gene expression, affecting diverse biological processes, notably programmed cell death (PCD). OBJECTIVE This review aims to explore the relationship between ncRNAs and PCD in CRC, focusing on how ncRNAs influence cancer cell survival, proliferation, and treatment resistance. METHODS A comprehensive literature analysis was conducted to examine recent findings on the role of ncRNAs in modulating various PCD mechanisms, including apoptosis, autophagy, necroptosis, and pyroptosis, and their impact on CRC development and therapeutic response. RESULTS ncRNAs were found to significantly regulate PCD pathways, impacting tumor growth, metastasis, and treatment sensitivity in CRC. Their influence on these pathways highlights the potential of ncRNAs as biomarkers for early CRC detection and as targets for innovative therapeutic interventions. CONCLUSION Understanding the involvement of ncRNAs in PCD regulation offers new insights into CRC biology. The targeted modulation of ncRNA-PCD interactions presents promising avenues for personalized cancer treatment, which may improve patient outcomes by enhancing therapeutic effectiveness and reducing resistance.
Collapse
Affiliation(s)
- Seyed Reza Taha
- Department of Pathology and Immunology, Washington University School of Medicine, St. LouisWashington, MO, USA
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kiev, Ukraine.
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Ali Bemani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahriar Kabirian
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Mohammadi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Jabbari
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Meysam Hushmand
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mokhtar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- PAKAN Institute, Tehran, Iran.
| |
Collapse
|
38
|
Zhang J, Zhang J, Yang C. Autophagy in brain tumors: molecular mechanisms, challenges, and therapeutic opportunities. J Transl Med 2025; 23:52. [PMID: 39806481 PMCID: PMC11727735 DOI: 10.1186/s12967-024-06063-0] [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: 07/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Autophagy is responsible for maintaining cellular balance and ensuring survival. Autophagy plays a crucial role in the development of diseases, particularly human cancers, with actions that can either promote survival or induce cell death. However, brain tumors contribute to high levels of both mortality and morbidity globally, with resistance to treatments being acquired due to genetic mutations and dysregulation of molecular mechanisms, among other factors. Hence, having knowledge of the role of molecular processes in the advancement of brain tumors is enlightening, and the current review specifically examines the role of autophagy. The discussion would focus on the molecular pathways that control autophagy in brain tumors, and its dual role as a tumor suppressor and a supporter of tumor survival. Autophagy can control the advancement of different types of brain tumors like glioblastoma, glioma, and ependymoma, demonstrating its potential for treatment. Autophagy mechanisms can influence metastasis and drug resistance in glioblastoma, and there is a complex interplay between autophagy and cellular responses to stress like hypoxia and starvation. Autophagy can inhibit the growth of brain tumors by promoting apoptosis. Hence, focusing on autophagy could offer fresh perspectives on creating successful treatments.
Collapse
Affiliation(s)
- Jiarui Zhang
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinan Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| |
Collapse
|
39
|
Hiruma K, Bilim V, Kazama A, Shirono Y, Murata M, Tomita Y. Acidic Microenvironment Enhances Cisplatin Resistance in Bladder Cancer via Bcl-2 and XIAP. Curr Issues Mol Biol 2025; 47:43. [PMID: 39852158 PMCID: PMC11763506 DOI: 10.3390/cimb47010043] [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: 12/23/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Cisplatin (CDDP) remains a key drug for patients with advanced bladder cancer (BC), despite the emergence of new therapeutic agents; thus, the identification of factors contributing to CDDP treatment resistance is crucial. As acidity of the tumor microenvironment has been reported to be associated with treatment resistance and poor prognosis across various cancer types, our objectives in this study were to investigate the effects of an acidic environment on BC cells and elucidate the mechanisms behind CDDP resistance. Our findings show that BC cells cultured under acidic conditions developed cisplatin resistance as acidity increased. Notably, CDDP administered to BC cells in a pH 6.0 environment required double the concentration, compared to those in a pH 7.5 environment, to achieve equivalent toxicity. Using chloroquine and navitoclax, we identified the involvement of the Bcl-2 and LC3B pathways in the acquisition of CDDP resistance under acidic conditions. A Western blot analysis revealed that the activations of Bcl-2 and XIAP expression appear to inhibit both apoptotic and autophagic cell death. Taken together, these results suggest that alleviating the acidity of the tumor microenvironment in clinical settings might enhance BC sensitivity to CDDP.
Collapse
Affiliation(s)
- Kaede Hiruma
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
| | - Vladimir Bilim
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Department of Urology, Kameda Daiichi Hospital, Niigata 950-0165, Japan
| | - Akira Kazama
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuko Shirono
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Department of Urology, Niigata Cancer Center Hospital, Niigata 951-8133, Japan
| | - Masaki Murata
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Department of Urology, Niigata Prefectural Central Hospital, Niigata 943-0192, Japan
| | - Yoshihiko Tomita
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
| |
Collapse
|
40
|
Loncke J, de Ridder I, Kale J, Wagner L, Kaasik A, Parys JB, Kerkhofs M, Andrews DW, Yule D, Vervliet T, Bultynck G. CISD2 counteracts the inhibition of ER-mitochondrial calcium transfer by anti-apoptotic BCL-2. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119857. [PMID: 39370046 DOI: 10.1016/j.bbamcr.2024.119857] [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: 05/09/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024]
Abstract
CISD2, a 2Fe2S cluster domain-containing protein, is implicated in Wolfram syndrome type 2, longevity and cancer. CISD2 is part of a ternary complex with IP3 receptors (IP3Rs) and anti-apoptotic BCL-2 proteins and enhances BCL-2's anti-autophagic function. Here, we examined how CISD2 impacted the function of BCL-2 in apoptosis and in controlling IP3R-mediated Ca2+ signaling. Using purified proteins, we found a direct interaction between the cytosolic region of CISD2 and BCL-2's BH4 domain with a submicromolar affinity. At the functional level, the cytosolic region of CISD2, as a purified protein, did not affect the ability of BCL-2 to inhibit BAX-pore formation. In a cellular context, loss of CISD2 did not impede the suppression of apoptosis by BCL-2. Also, in Ca2+-signaling assays, absence of CISD2 did not affect the inhibition of IP3R-mediated Ca2+ release by BCL-2. Combined, these experiments indicate that CISD2 is not essential for BCL-2 function in apoptosis and cytosolic Ca2+ signaling. Instead, CISD2 overexpression enhanced BCL-2-mediated suppression of cytosolic IP3R-mediated Ca2+ release. However, consistent with the presence of CISD2 and BCL-2 at mitochondria-associated ER membranes (MAMs), the most striking effect was observed at the level of ER-mitochondrial Ca2+ transfer. While BCL-2 overexpression inhibited ER-mitochondrial Ca2+ transfer, overexpression of CISD2 together with BCL-2 abrogated the effect of BCL-2. The underlying mechanism is linked to ER-mitochondrial contact sites, since BCL-2 reduced ER-mitochondrial contact sites while co-expression of CISD2 together with BCL-2 abolished this effect. These findings reveal a unique interplay between BCL-2 and CISD2 at Ca2+-signaling nanodomains between ER and mitochondria.
Collapse
Affiliation(s)
- Jens Loncke
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Ian de Ridder
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Justin Kale
- University of Toronto, Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Larry Wagner
- University of Rochester, Department of Pharmacology and Physiology, School of Medicine and Dentistry, 601 Elmwood Avenue, Box 711, Rochester, NY 14642, USA
| | - Allen Kaasik
- University of Tartu, Department of Pharmacology, Institute of Biomedicine and Translational Medicine, Tartu, Estonia
| | - Jan B Parys
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Martijn Kerkhofs
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium; Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008 Lyon, France
| | - David W Andrews
- University of Toronto, Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - David Yule
- University of Rochester, Department of Pharmacology and Physiology, School of Medicine and Dentistry, 601 Elmwood Avenue, Box 711, Rochester, NY 14642, USA
| | - Tim Vervliet
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium.
| |
Collapse
|
41
|
Larrañaga-SanMiguel A, Bengoa-Vergniory N, Flores-Romero H. Crosstalk between mitochondria-ER contact sites and the apoptotic machinery as a novel health meter. Trends Cell Biol 2025; 35:33-45. [PMID: 39379268 DOI: 10.1016/j.tcb.2024.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024]
Abstract
Mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) function as transient signaling platforms that regulate essential cellular functions. MERCS are enriched in specific proteins and lipids that connect mitochondria and the ER together and modulate their activities. Dysregulation of MERCS is associated with several human pathologies including Alzheimer's disease (AD), Parkinson's disease (PD), and cancer. BCL-2 family proteins can locate at MERCS and control essential cellular functions such as calcium signaling and autophagy in addition to their role in mitochondrial apoptosis. Moreover, the BCL-2-mediated apoptotic machinery was recently found to trigger cGAS-STING pathway activation and a proinflammatory response, a recognized hallmark of these diseases that requires mitochondria-ER interplay. This review underscores the pivotal role of MERCS in regulating essential cellular functions, focusing on their crosstalk with BCL-2 family proteins, and discusses how their dysregulation is linked to disease.
Collapse
Affiliation(s)
| | - Nora Bengoa-Vergniory
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy, and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Hector Flores-Romero
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
42
|
Nunes-Pinto M, Bandeira de Mello RG, Pinto MN, Moro C, Vellas B, Martinez LO, Rolland Y, de Souto Barreto P. Sarcopenia and the biological determinants of aging: A narrative review from a geroscience perspective. Ageing Res Rev 2025; 103:102587. [PMID: 39571617 DOI: 10.1016/j.arr.2024.102587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND The physiopathology of sarcopenia shares common biological cascades with the aging process, as does any other age-related condition. However, our understanding of the interconnected pathways between diagnosed sarcopenia and aging remains limited, lacking sufficient scientific evidence. METHODS This narrative review aims to gather and describe the current evidence on the relationship between biological aging determinants, commonly referred to as the hallmarks of aging, and diagnosed sarcopenia in humans. RESULTS Among the twelve hallmarks of aging studied, there appears to be a substantial association between sarcopenia and mitochondrial dysfunction, epigenetic alterations, deregulated nutrient sensing, and altered intercellular communication. Although limited, preliminary evidence suggests a promising association between sarcopenia and genomic instability or stem cell exhaustion. DISCUSSION Overall, an imbalance in energy regulation, characterized by impaired mitochondrial energy production and alterations in circulatory markers, is commonly associated with sarcopenia and may reflect the interplay between aging physiology and sarcopenia biology.
Collapse
Affiliation(s)
- Mariá Nunes-Pinto
- Gerontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France; Postgraduate Program in Medical Sciences (Endocrinology), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| | - Renato Gorga Bandeira de Mello
- Postgraduate Program in Medical Sciences (Endocrinology), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; School of Medicine, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Master of Public Health Program, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, USA
| | - Milena Nunes Pinto
- School of Medicine, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Cédric Moro
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), Toulouse UMR1297, France
| | - Bruno Vellas
- Gerontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France; CERPOP UMR 1295, University of Toulouse III, Institut National de la Santé et de la Recherche Médicale (Inserm), UPS, Toulouse, France; IHU HealthAge, Toulouse, France
| | - Laurent O Martinez
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), Toulouse UMR1297, France; IHU HealthAge, Toulouse, France
| | - Yves Rolland
- Gerontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France; CERPOP UMR 1295, University of Toulouse III, Institut National de la Santé et de la Recherche Médicale (Inserm), UPS, Toulouse, France; IHU HealthAge, Toulouse, France
| | - Philipe de Souto Barreto
- Gerontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France; CERPOP UMR 1295, University of Toulouse III, Institut National de la Santé et de la Recherche Médicale (Inserm), UPS, Toulouse, France; IHU HealthAge, Toulouse, France
| |
Collapse
|
43
|
Jahani M, Yarani R, Rezazadeh D, Tahmasebi H, Hoseinkhani Z, Kiani S, Mansouri K. L-lysine Increases the Anticancer Effect of Doxorubicin in Breast Cancer by Inducing ROS-dependent Autophagy. Curr Cancer Drug Targets 2025; 25:257-269. [PMID: 38584530 DOI: 10.2174/0115680096288665240315072646] [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: 12/07/2023] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Doxorubicin (DOX) is a chemotherapy drug that is widely used in cancer therapy, especially in Triple-Negative Breast Cancer (TNBC) patients. Nevertheless, cytoprotective autophagy induction by DOX limits its cytotoxic effect and drug resistance induction in patients. Therefore, finding a new way is essential for increasing the effectiveness of this drug for cancer treatment. OBJECTIVE This study aimed to investigate the effect of L-lysine on DOX cytotoxicity, probably through autophagy modulation in TNBC cell lines. METHODS We used two TNBC cell lines, MDA-MB-231 and MDA-MB-468, with various levels of autophagy activity. Cell viability after treatment with L-lysine alone and in combination therapy was evaluated by MTT assay. Reactive Oxygen Species (ROS), nitric oxide (NO) concentration, and arginase activity were assessed using flow cytometric analysis, Griess reaction, and arginase activity assay kit, respectively. Real-time PCR and western blot analysis were used to evaluate the L-lysine effect on the autophagy-related genes and protein expression. Cell cycle profile and apoptotic assay were performed using flow cytometric analysis. RESULTS The obtained data indicated that L-lysine in both concentrations of 24 and 32 mM increased the autophagy flux and enhanced the DOX cytotoxicity, especially in MDA-MB-231, which demonstrated higher autophagy activity than MDA-MB-468, by inducing ROS and NO production. Furthermore, L-lysine induced G2/M arrest autophagy cell death, while significant apoptotic changes were not observed. CONCLUSION These findings suggest that L-lysine can increase DOX cytotoxicity through autophagy modulation. Thus, L-lysine, in combination with DOX, may facilitate the development of novel adjunct therapy for cancer.
Collapse
Affiliation(s)
- Mozhgan Jahani
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Yarani
- Steno Diabetes Center Copenhagen, The Capital Region of Denmark Pediatrics, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Davood Rezazadeh
- Department of Molecular Medicine, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hadis Tahmasebi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Hoseinkhani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sara Kiani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Department of Molecular Medicine, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
44
|
Wilson A, McCormick C. Reticulophagy and viral infection. Autophagy 2025; 21:3-20. [PMID: 39394962 DOI: 10.1080/15548627.2024.2414424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/14/2024] Open
Abstract
All viruses are obligate intracellular parasites that use host machinery to synthesize viral proteins. In infected eukaryotes, viral secreted and transmembrane proteins are synthesized at the endoplasmic reticulum (ER). Many viruses refashion ER membranes into bespoke factories where viral products accumulate while evading host pattern recognition receptors. ER processes are tightly regulated to maintain cellular homeostasis, so viruses must either conform to ER regulatory mechanisms or subvert them to ensure efficient viral replication. Reticulophagy is a catabolic process that directs lysosomal degradation of ER components. There is accumulating evidence that reticulophagy serves as a form of antiviral defense; we call this defense "xERophagy" to acknowledge its relationship to xenophagy, the catabolic degradation of microorganisms by macroautophagy/autophagy. In turn, viruses can subvert reticulophagy to suppress host antiviral responses and support efficient viral replication. Here, we review the evidence for functional interplay between viruses and the host reticulophagy machinery.Abbreviations: AMFR: autocrine motility factor receptor; ARF4: ADP-ribosylation factor 4; ARL6IP1: ADP-ribosylation factor-like 6 interacting protein 1; ATL3: atlastin GTPase 3; ATF4: activating transcription factor 4; ATF6: activating transcription factor 6; BPIFB3: BPI fold containing family B, member 3; CALCOCO1: calcium binding and coiled coil domain 1; CAMK2B: calcium/calmodulin-dependent protein kinase II, beta; CANX: calnexin; CDV: canine distemper virus; CCPG1: cell cycle progression 1; CDK5RAP3/C53: CDK5 regulatory subunit associated protein 3; CIR: cargo-interacting region; CoV: coronavirus; CSNK2/CK2: casein kinase 2; CVB3: coxsackievirus B3; DAPK1: death associated protein kinase 1; DENV: dengue virus; DMV: double-membrane vesicles; EBOV: Ebola virus; EBV: Epstein-Barr Virus; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; EMCV: encephalomyocarditis virus; EMV: extracellular microvesicle; ER: endoplasmic reticulum; ERAD: ER-associated degradation; ERN1/IRE1: endoplasmic reticulum to nucleus signalling 1; EV: extracellular vesicle; EV71: enterovirus 71; FIR: RB1CC1/FIP200-interacting region; FMDV: foot-and-mouth disease virus; HCMV: human cytomegalovirus; HCV: hepatitis C virus; HMGB1: high mobility group box 1; HSPA5/BiP: heat shock protein 5; IFN: interferon; IFNG/IFN-γ: interferon gamma; KSHV: Kaposi's sarcoma-associated herpesvirus; LIR: MAP1LC3/LC3-interacting region; LNP: lunapark, ER junction formation factor; MAP1LC3: microtubule-associated protein 1 light chain 3; MAP3K5/ASK1: mitogen-activated protein kinase kinase kinase 5; MAPK/JNK: mitogen-activated protein kinase; MeV: measles virus; MHV: murine hepatitis virus; NS: non-structural; PDIA3: protein disulfide isomerase associated 3; PRR: pattern recognition receptor; PRRSV: porcine reproductive and respiratory syndrome virus; RB1CC1/FIP200: RB1-inducible coiled-coil 1; RETREG1/FAM134B: reticulophagy regulator 1; RHD: reticulon homology domain; RTN3: reticulon 3; RTN3L: reticulon 3 long; sAIMs: shuffled Atg8-interacting motifs; SARS-CoV: severe acute respiratory syndrome coronavirus; SINV: Sindbis virus; STING1: stimulator of interferon response cGAMP interactor 1; SVV: Seneca Valley virus; SV40: simian virus 40; TEX264: testis expressed gene 264 ER-phagy receptor; TFEB: transcription factor EB; TRAF2: TNF receptor-associated factor 2; UIM: ubiquitin-interacting motif; UFM1: ubiquitin-fold modifier 1; UPR: unfolded protein response; VAPA: vesicle-associated membrane protein, associated protein A; VAPB: vesicle-associated membrane protein, associated protein B and C; VZV: varicella zoster virus; WNV: West Nile virus; XBP1: X-box binding protein 1; XBP1s: XBP1 spliced; xERophagy: xenophagy involving reticulophagy; ZIKV: Zika virus.
Collapse
Affiliation(s)
- Alexa Wilson
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Craig McCormick
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
45
|
Alimoradi N, Ramezani A, Tahami M, Firouzabadi N. Metformin Exhibits Anti-Inflammatory Effects by Regulating microRNA-451/CXCL16 and B Cell Leukemia/Lymphoma 2 in Patients With Osteoarthritis. ACR Open Rheumatol 2025; 7:e11755. [PMID: 39435687 PMCID: PMC11694140 DOI: 10.1002/acr2.11755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is the most common cause of chronic disability in joints among older individuals. The primary goal of OA treatment is pain relief to improve the quality of life. Inflammation and aging are involved in the pathogenesis of pain in OA. In this study, we evaluated the ability of metformin to regulate microRNAs, such as miR-451 and miR-15b, and their target proteins, CXCL16 and B cell leukemia/lymphoma 2 (BCL-2), involved in inflammation and apoptosis. METHODS In this double-blind placebo-controlled clinical trial, patients were randomly divided into two groups: one receiving metformin and the other receiving a placebo for four months (starting at 0.5 g/day for the first week, increasing to 1 g/day for the second week, and increasing to 1.5 g/day for the remaining period). In addition to evaluating the clinical response using the Knee Injury and Osteoarthritis Outcome Score questionnaire, miR-451 and miR-15b expression levels were detected using real-time polymerase chain reaction. The serum levels of CXCL16 and BCL-2 were evaluated using enzyme-linked immunosorbent assay kits before (time zero) and after treatment (month four). RESULTS Metformin increased miR-451 expression levels simultaneously with pain reduction, whereas miR-15b expression did not change significantly after four months of treatment. Also, metformin decreased the serum levels of BCL-2 and CXCL16 in patients with OA. CONCLUSION The effects of metformin in reducing pain can be attributed to many factors, including its anti-inflammatory and antiaging effects. Our findings suggest that metformin may reduce pain and inflammation in patients with OA through the regulation of miR-451/CXCL16 and BCL-2.
Collapse
|
46
|
Liu Y, Qin K, Dou K, Ren J, Hou B, Xie A. TMEM106B knockdown exhibits a neuroprotective effect in Parkinson's disease models via regulating autophagy-lysosome pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167553. [PMID: 39490939 DOI: 10.1016/j.bbadis.2024.167553] [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: 06/12/2024] [Revised: 09/14/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND TMEM106B, a lysosomal transmembrane protein, has been reported to be associated with Parkinson's disease (PD). However, the precise physiopathologic mechanism of TMEM106B in PD remains unclear. OBJECTIVE This study aimed to explore the influence of TMEM106B on the autophagy-lysosome pathway (ALP) in PD. METHODS 55 patients with PD and 40 healthy controls were enrolled. RT-qPCR and ELISA were employed to assess the levels of TMEM106B. In vitro and in vivo models of PD, Lentivirus-shTMEM106B and AAV-shTMEM106B were used to knockdown the expression of TMEM106B. Behavioral experiments, western blot, immunofluorescence, and immunohistochemistry were used to detect the effect of TMEM106B on the ALP process. RESULTS We found that the levels of TMEM106B were increased in the PD patients and PD models. TMEM106B knockdown markedly improved the motor deficits and tyrosine hydroxylase (TH) expression of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned mice. TMEM106B knockdown promoted α-syn clearance by regulating the ALP process in MPP+-induced SH-SY5Y cells and MPTP-treated mice. Further studies revealed that TMEM106B knockdown might activate ALP through activating AMPK-mTOR-TFEB axis. Furthermore, TMEM106B may play a vital role in the ALP by mediating the expression of TDP43. CONCLUSIONS Taken together, our study suggests that TMEM106B knockdown mediates the ALP pathway, leading to a decrease in α-syn, providing a new direction and perspective for the regulation of autophagy in PD.
Collapse
Affiliation(s)
- Yumei Liu
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kunpeng Qin
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kaixin Dou
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiwen Ren
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Binghui Hou
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
47
|
Schmidt KW, Montespan C, Thompson D, Lucas MS, Ligeon LA, Wodrich H, Hahn AS, Greber UF, Münz C. Selective autophagy impedes KSHV entry after recruiting the membrane damage sensor galectin-8 to virus-containing endosomes. Cell Rep 2024; 43:115019. [PMID: 39602307 DOI: 10.1016/j.celrep.2024.115019] [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: 11/30/2023] [Revised: 10/15/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is an oncogenic γ-herpesvirus. Autophagy during KSHV entry has remained unexplored. We show that LC3 lipidation as a hallmark of autophagy is induced shortly after KSHV entry. LC3 co-localizes with KSHV in amphisomes during entry and loss of LC3 lipidation increases infection. Accordingly, NDP52, a receptor of selective autophagy, was recruited to endocytosed viral particles, and its reduction increased KSHV infection. Additionally, virus particles co-localized with the endolysosome damage sensor galectin-8 upon KSHV entry and depletion of galectin-8 promoted KSHV infection. Compared with herpes simplex virus, listeriolysin, adenovirus, and influenza virus, and in contrast to what was previously thought about enveloped viruses, KSHV binding to EphA2 by its envelope protein gH causes endolysosomal membrane damage, akin to non-enveloped viruses and bacteria. Taken together, our study identifies an important anti-viral role for galectin-8, NDP52, and the autophagy machinery at virus-damaged endosomes, restricting KSHV entry by selective autophagy.
Collapse
Affiliation(s)
- Katarina Wendy Schmidt
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Charlotte Montespan
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Danielle Thompson
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Miriam S Lucas
- ScopeM - Scientific Center for Optical and Electron Microscopy, ETH Zurich, 8093 Zurich, Switzerland
| | - Laure-Anne Ligeon
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Harald Wodrich
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, University of Bordeaux, 33063 Bordeaux, France
| | - Alexander S Hahn
- German Primate Center, University of Göttingen, 37077 Göttingen, Germany
| | - Urs F Greber
- Institute of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
48
|
Andrei C, Mihai DP, Nitulescu GM, Nitulescu G, Zanfirescu A. Modulating Autophagy in Osteoarthritis: Exploring Emerging Therapeutic Drug Targets. Int J Mol Sci 2024; 25:13695. [PMID: 39769455 PMCID: PMC11727697 DOI: 10.3390/ijms252413695] [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: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by the breakdown of cartilage and the subsequent inflammation of joint tissues, leading to pain and reduced mobility. Despite advancements in symptomatic treatments, disease-modifying therapies for OA remain limited. This narrative review examines the dual role of autophagy in OA, emphasizing its protective functions during the early stages and its potential to contribute to cartilage degeneration in later stages. By delving into the molecular pathways that regulate autophagy, this review highlights its intricate interplay with oxidative stress and inflammation, key drivers of OA progression. Emerging therapeutic strategies aimed at modulating autophagy are explored, including pharmacological agents such as AMP kinase activators, and microRNA-based therapies. Preclinical studies reveal encouraging results, demonstrating that enhancing autophagy can reduce inflammation and decelerate cartilage degradation. However, the therapeutic benefits of autophagy modulation depend on precise, stage-specific approaches. Excessive or dysregulated autophagy in advanced OA may lead to chondrocyte apoptosis, exacerbating joint damage. This review underscores the promise of autophagy-based interventions in bridging the gap between experimental research and clinical application. By advancing our understanding of autophagy's role in OA, these findings pave the way for innovative and effective therapies. Nonetheless, further research is essential to optimize these strategies, address potential off-target effects, and develop safe, targeted treatments that improve outcomes for OA patients.
Collapse
Affiliation(s)
| | - Dragos Paul Mihai
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.A.); (G.M.N.); (G.N.); (A.Z.)
| | | | | | | |
Collapse
|
49
|
Vervier J, Squatrito M, Nisolle M, Henry L, Munaut C. Controversial Roles of Autophagy in Adenomyosis and Its Implications for Fertility Outcomes-A Systematic Review. J Clin Med 2024; 13:7501. [PMID: 39768424 PMCID: PMC11676161 DOI: 10.3390/jcm13247501] [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: 11/07/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Adenomyosis is a benign condition where ectopic endometrial glandular tissue is found within the uterine myometrium. Its impact on women's reproductive outcomes is substantial, primarily due to defective decidualization, impaired endometrial receptivity, and implantation failure. The exact pathogenesis of the disease remains unclear, and the role of autophagy in adenomyosis and its associated infertility is not well understood. The aim of this systematic review was to conduct an exhaustive search of the literature to clarify the role of autophagy in the pathogenesis of adenomyosis. Methods: A systematic search was conducted in Medline, Embase, and Scopus databases up to the date of 20 August 2024. We included all English-written publications assessing the role of autophagy in the pathogenesis of adenomyosis. Results: Seventeen eligible articles were identified, including reviews and experimental studies involving human samples and murine models. The results showed that the role of autophagy in adenomyosis is controversial, with studies showing both increased and decreased levels of autophagy in adenomyosis. Conclusions: Autophagy plays a dual role in cell survival and death. Increased autophagy might support the survival and proliferation of ectopic endometrial cells, while decreased autophagy could prevent cell death, leading to abnormal growth. Oxidative stress may trigger pro-survival autophagy, mitigating apoptosis and promoting cellular homeostasis. Hormonal imbalances disrupt normal autophagic activity, potentially impairing endometrial receptivity and decidualization and contributing to infertility. The balance of autophagy is crucial in adenomyosis, with its dual role contributing to the complexity of the disease. Limitations: A few studies have been conducted with heterogeneous populations, limiting comparative analyses.
Collapse
Affiliation(s)
- Julie Vervier
- Laboratory of Tumor and Development Biology, Giga-Cancer, University of Liège, 4000 Liege, Belgium
- Obstetrics and Gynecology Department, University of Liège-Citadelle Site, 4000 Liege, Belgium
| | - Marlyne Squatrito
- Laboratory of Tumor and Development Biology, Giga-Cancer, University of Liège, 4000 Liege, Belgium
| | - Michelle Nisolle
- Obstetrics and Gynecology Department, University of Liège-Citadelle Site, 4000 Liege, Belgium
| | - Laurie Henry
- Obstetrics and Gynecology Department, University of Liège-Citadelle Site, 4000 Liege, Belgium
- Center for Reproductive Medicine, University of Liège-Citadelle Site, 4000 Liege, Belgium
| | - Carine Munaut
- Laboratory of Tumor and Development Biology, Giga-Cancer, University of Liège, 4000 Liege, Belgium
| |
Collapse
|
50
|
Li Y, Bel S, Benjamin JL, Ruhn KA, Hassell B, Behrendt CL, Kuang Z, Hooper LV. BCL2 regulates antibacterial autophagy in the intestinal epithelium. Proc Natl Acad Sci U S A 2024; 121:e2410205121. [PMID: 39602254 PMCID: PMC11626146 DOI: 10.1073/pnas.2410205121] [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: 06/03/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Autophagy is a key innate immune defense mechanism in intestinal epithelial cells. Bacterial invasion of epithelial cells activates antibacterial autophagy through a process that requires the innate immune adaptor protein MYD88, yet how MYD88 signaling connects to the autophagy machinery is unknown. Here, we show that the mouse intestinal pathogen Salmonella enterica Serovar Typhimurium (Salmonella Typhimurium) triggers MYD88 signaling that regulates binding of the anti-autophagy factor B cell lymphoma 2 (BCL2) to the essential autophagy protein Beclin1 (BECN1) in small intestinal enterocytes, a key epithelial cell lineage. Salmonella infection activated the kinase c-Jun N-terminal protein kinase 1 (JNK1) downstream of MYD88. JNK1 induced enterocyte BCL2 phosphorylation, promoting dissociation of the inhibitory BCL2-BECN1 complex and releasing BECN1 to initiate autophagy. Mice with BCL2 phosphorylation site mutations that prevent BCL2-BECN1 dissociation showed increased Salmonella invasion of enterocytes and dissemination to extraintestinal sites. These findings reveal that BCL2 links MYD88 signaling to enterocyte autophagy initiation, providing mechanistic insight into how invading bacteria trigger autophagy in the intestinal epithelium.
Collapse
Affiliation(s)
- Yun Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Shai Bel
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Jamaal L. Benjamin
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Kelly A. Ruhn
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Brian Hassell
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Cassie L. Behrendt
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Zheng Kuang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Lora V. Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX75390
- HHMI, University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|