1
|
Bao C, Zhang Y, Feng J, Hong X, Gao N, Feng G. Deciphering tuberculosis: lysosome-centric insights into pathogenesis and therapies. Front Cell Infect Microbiol 2025; 15:1582037. [PMID: 40438237 PMCID: PMC12116394 DOI: 10.3389/fcimb.2025.1582037] [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: 02/23/2025] [Accepted: 04/17/2025] [Indexed: 06/01/2025] Open
Abstract
Tuberculosis is a widely spread disease caused by Mycobacterium tuberculosis (Mtb). The pathogenicity of the pathogen is closely associated with the immune defense mechanisms of the host cells. As key cellular degradation and metabolic centers, lysosomes critically regulate tuberculosis infection. When Mtb invades the host, it is taken up by macrophages and enters phagosomes. Subsequently, the phagosomes fuse with lysosomes and form phagolysosomes, which eliminate the pathogenic bacteria through the acidic environment and hydrolytic enzymes within lysosomes. However, Mtb can interfere with the normal functions of lysosomes through various strategies. It can secrete specific factors (such as ESAT-6, ppk-1, and AcpM) to inhibit the acidification of lysosomes, enzyme activity, and the fusion of phagosomes and lysosomes, thereby enabling Mtb proliferation within host cells. An in-depth exploration of the mechanism of the interaction between Mtb and lysosomes will both uncover bacterial immune evasion strategies and identify novel anti-tuberculosis therapeutic targets.
Collapse
Affiliation(s)
- Cui Bao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuanyuan Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiao Feng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiuwen Hong
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nan Gao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ganzhu Feng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Zhang W, Ji C, Li X, He T, Jiang W, Liu Y, Wu M, Zhao Y, Chen X, Wang X, Li J, Zhang H, Wang J. Autophagy-independent role of ATG9A vesicles as carriers for galectin-9 secretion. Nat Commun 2025; 16:4259. [PMID: 40335523 PMCID: PMC12059159 DOI: 10.1038/s41467-025-59605-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
Galectins play vital roles in cellular processes such as adhesion, communication, and survival, yet the mechanisms underlying their unconventional secretion remain poorly understood. This study identifies ATG9A, a core autophagy protein, as a key regulator of galectin-9 secretion via a mechanism independent of classical autophagy, secretory autophagy, or the LC3-dependent extracellular vesicle loading and secretion pathway. ATG9A vesicles function as specialized carriers, with the N-terminus of ATG9A and both carbohydrate recognition domains of galectin-9 being critical for the process. TMED10 mediates the incorporation of galectin-9 into ATG9A vesicles, which then fuse with the plasma membrane via the STX13-SNAP23-VAMP3 SNARE complex. Furthermore, ATG9A regulates the secretion of other proteins, including galectin-4, galectin-8, and annexin A6, but not IL-1β, galectin-3, or FGF2. This mechanism is potentially conserved across other cell types, including monocytic cells, which underscores its broader significance in unconventional protein secretion.
Collapse
Affiliation(s)
- Wenting Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Cuicui Ji
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xianghua Li
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Tianlong He
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Wei Jiang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Yukun Liu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Meiling Wu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Yunpeng Zhao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xuechai Chen
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xiaoli Wang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Jian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Haolin Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China.
| | - Juan Wang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China.
| |
Collapse
|
3
|
Ulecia-Morón C, Bris ÁG, MacDowell KS, Cerveró-García P, Madrigal JLM, García-Bueno B, Pereira MP, Leza JC, Caso JR. Chronic mild stress dysregulates autophagy, membrane dynamics, and lysosomal status in frontal cortex and hippocampus of rats. Eur Neuropsychopharmacol 2025; 94:24-35. [PMID: 40056662 DOI: 10.1016/j.euroneuro.2025.02.005] [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: 07/19/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/10/2025]
Abstract
Inflammation has been related to major depressive disorder pathophysiology. Autophagy, a degradative pathway regulating inflammation and immunity, has emerged as a potential contributor. Among others, we characterized, in frontal cortex (FC) and hippocampus (Hp), autophagy markers (upregulations in mTOR, ATG7, and ATG 16L1, and downregulations in ULK1, BECLIN1, phospho-SQSTM1, ATG3, ATG12, and ATG 16L1), effectors of the endosomal sorting complexes required for transport (overexpression in HRS, VPS37A, CHMP6, and GALECTIN 3, and downregulations in STAM2, TSG101, VPS28, VPS37A, CHMP5, VPS4B, and GALECTIN 9), and lysosomal proteins (LAMP1, LAMP2A, MANNOSE RECEPTOR, HSC70, HSP70, CATHEPSIN D and B, and CYSTATIN C, whose variations are dependent on lysosomal nature and brain region) of male rats exposed to chronic mild stress, a model of depression, compared to control rats. Results indicate that chronic stress alters protein expression of autophagy and the endosomal sorting complexes required for transport markers in a region-specific manner, plus increases lysosomal presence, oppositely modulating lysosomal proteins in each structure. Additionally, astrocytes seemed to exert an essential role in the regulation of the autophagy adaptor SQSTM1/p62. In conclusion, stress-induced protein disruptions in these pathways highlight their differential modulation after chronic stress exposure and their potential role in maintaining brain homeostasis during the stress response, making them promising targets for new therapeutic strategies in stress-related pathologies.
Collapse
Affiliation(s)
- Cristina Ulecia-Morón
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación Neuroquímica (IUIN, UCM), Madrid, Spain
| | - Álvaro G Bris
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación Neuroquímica (IUIN, UCM), Madrid, Spain
| | - Karina S MacDowell
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación Neuroquímica (IUIN, UCM), Madrid, Spain
| | - Pilar Cerveró-García
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Neurociencias de Castilla y León (INCyL), Salamanca, Spain
| | - José L M Madrigal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación Neuroquímica (IUIN, UCM), Madrid, Spain
| | - Borja García-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación Neuroquímica (IUIN, UCM), Madrid, Spain
| | - Marta P Pereira
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Centro de Biología Molecular "Severo Ochoa" (CBMSO, UAM-CSIC), Instituto Universitario de Biología Molecular (IUBM-UAM), Madrid, Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación Neuroquímica (IUIN, UCM), Madrid, Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Instituto Universitario de Investigación Neuroquímica (IUIN, UCM), Madrid, Spain.
| |
Collapse
|
4
|
Abooali M, Schlichtner S, Lei X, Aliu N, Ruggiero S, Loges S, Ziegler M, Hertel F, Volckmar AL, Stenzinger A, Christopoulos P, Thomas M, Klenova E, Meyer NH, Boussios S, Heaton N, Zen Y, Zamalloa A, Chokshi S, Urbani L, Richard S, Kirubendran K, Hussain R, Siligardi G, Cholewa D, Berger SM, Yasinska IM, Fasler-Kan E, Sumbayev VV. Intracellular and extracellular activities of V-domain Ig-containing suppressor of T cell activation (VISTA) modulated by immunosuppressive factors of tumour microenvironment. Cancer Lett 2025; 616:217581. [PMID: 39983894 DOI: 10.1016/j.canlet.2025.217581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/23/2025]
Abstract
V-domain Ig-containing suppressor of T cell activation (VISTA) is a unique immune checkpoint protein, which was reported to display both receptor and ligand activities. However, the mechanisms of regulation of VISTA activity and functions by factors of tumour microenvironment (TME) remain unclear and understanding these processes is required in order to develop successful personalised cancer immunotherapeutic strategies and approaches. Here we report for the very first time that VISTA interacts with another immune checkpoint protein galectin-9 inside the cell most likely facilitating its interaction with TGF-β-activated kinase 1 (TAK1). This process is required for protection of lysosomes, which is crucial for many cell types and tissues. We found that VISTA expression can be differentially controlled by crucial factors present in TME, such as transforming growth factor beta type 1 (TGF-β) and hypoxia as well as other factors activating hypoxic signalling. We confirmed that involvement of these important pathways modulated by TME differentially influences VISTA expression in different cell types. These networks include: TGF-β-Smad3 pathway, TAK1 (TGF-β-activated kinase 1) or apoptosis signal-regulating kinase 1 (ASK1)-induced activation of activating transcription factor 2 (ATF-2) and hypoxic signalling pathway. Based on this work we determined the five critical functions of VISTA and the role of TME factors in controlling (modulating or downregulating) VISTA expression.
Collapse
Affiliation(s)
- Maryam Abooali
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Stephanie Schlichtner
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Xi Lei
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Nijas Aliu
- Department of Human Genetics, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Sonia Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Ziegler
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Franziska Hertel
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Elena Klenova
- School of Life Sciences, University of Essex, Colchester, UK
| | - N Helge Meyer
- Division of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, CT1 1QU, Kent, UK; Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King's College London, Strand, London, WC2R 2LS, UK; Kent Medway Medical School, University of Kent, Canterbury, CT2 7LX, Kent, UK; Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, ME7 5NY, Kent, UK; AELIA Organization, 9th Km Thessaloniki-Thermi, 57001, Thessaloniki, Greece
| | - Nigel Heaton
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Yoh Zen
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Ane Zamalloa
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Shilpa Chokshi
- Peninsula Medical School, Faculty of Health, University of Plymouth, UK; Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Luca Urbani
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Sophie Richard
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Kavitha Kirubendran
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | | | | | - Dietmar Cholewa
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland.
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK.
| |
Collapse
|
5
|
Tian N, Chu H, Li Q, Sun H, Zhang J, Chu N, Sun Z. Host-directed therapy for tuberculosis. Eur J Med Res 2025; 30:267. [PMID: 40211397 PMCID: PMC11987284 DOI: 10.1186/s40001-025-02443-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/09/2025] [Indexed: 04/13/2025] Open
Abstract
Current TB treatment regimens are hindered by drug resistance, numerous adverse effects, and long treatment durations, highlighting the need for 'me-better' treatment regimens. Host-directed therapy (HDT) has gained recognition as a promising approach in TB treatment. It allows the repurposing of existing drugs approved for other conditions and aims to enhance the effectiveness of existing anti-TB therapies, minimize drug resistance, decrease treatment duration, and adverse effects. By modulating the host immune response, HDT ameliorates immunopathological damage and improves overall outcomes by promoting autophagy, antimicrobial peptide production, and other mechanisms. It holds promise for addressing the challenges posed by multiple and extensively drug-resistant Mycobacterium tuberculosis strains, which are increasingly difficult to treat using conventional therapies. This article reviews various HDT candidates, including repurposed drugs, explores their underlying mechanisms such as autophagy promotion and inflammation reduction, while emphasizing their potential to improve TB treatment outcomes and outlining future research directions.
Collapse
Affiliation(s)
- Na Tian
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Qi Li
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Hong Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jingfang Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Naihui Chu
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
6
|
Srivastava SP, Kopasz-Gemmen O, Kunamneni A, Thurnman A, Ozukan E, Swaroop V, Yoshida S, Hong S, Inoki K. AMPK is dispensable for physiological podocyte and glomerular functions but prevents glomerular fibrosis in experimental diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647592. [PMID: 40291739 PMCID: PMC12026990 DOI: 10.1101/2025.04.07.647592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
AMP-activated protein kinase (AMPK) has been postulated to be crucial in regulating various renal physiology and pathophysiology processes, including energy metabolism, ion and water transport, inflammation, and hypertrophy. However, the specific roles of AMPK in the podocyte, a cell critical for maintaining glomerular filtration, have not been fully explored using genetic model animals. In this study, we generated mice lacking both AMPK α1 and α2 catalytic subunits in glomerular podocytes (pmut). Our findings revealed that, surprisingly, AMPK is dispensable for normal podocyte function. These knockout mice could live as long as their wild-type littermates without showing any pathological alterations in their glomeruli or glomerular function at two years of age. However, under type 1 diabetic conditions, the diabetic pmut mice exhibited increased lipid and collagen accumulation and an elevated expression of mesenchymal proteins in their glomeruli. They also showed more significant albuminuria compared to control diabetic mice. Under high glucose culture conditions, glomeruli isolated from pmut mice demonstrated a reduced expression of mitochondrial genes (e.g., Ndufv2) and increased leakage of mitochondrial components. Additionally, there was heightened expression of genes associated with nucleotide sensing and pro-inflammatory pathways (including mb21d2, IL-1 beta, and NF-kB). These observations suggest that while AMPK is not necessary for podocyte function in healthy kidneys, it is crucial for preventing glomerular fibrosis resulting from lipotoxicity and inflammation under diabetic conditions.
Collapse
|
7
|
Xu J, He X, Zhang S, Li L, Li P. Expression of co-signaling molecules TIM-3/Galectin-9 at the maternal-fetal interface. Placenta 2025; 163:43-50. [PMID: 40068377 DOI: 10.1016/j.placenta.2025.03.002] [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: 11/26/2024] [Revised: 01/31/2025] [Accepted: 03/02/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION During early pregnancy, fetal placental tissue implants into maternal decidual tissue, forming a unique interface where maternal immune cells do not reject the invading fetal cells. Given the roles of Galectin-9 and Tim-3 in tumor immune regulation, studying their distribution and function at this interface may provide insights into recurrent pregnancy loss. METHODS This study uses single-cell transcriptomics, spatial transcriptomics, and multiplex immunohistochemistry to examine the expression and localization of Galectin-9 and TIM-3. Hormone-induced decidualization of immortalized human endometrial stromal cells was conducted to investigate Galectin-9 expression. RESULTS The major immune cells in the maternal decidua, such as T cells, NK cells, and macrophages, co-express Galectin-9 and TIM-3. Unlike TIM-3, Galectin-9 is also highly expressed in endothelial cells and decidualized stromal cells. Among placenta-derived cells, Hofbauer cells (HBs) and Placenta-associated maternal monocytes/macrophages (PAMMs) exhibit high expression of both Galectin-9 and TIM-3, while trophoblast cells show relatively low levels of expression. Additionally, hormone-induced decidualization significantly upregulates Galectin-9 expression in endometrial stromal cells. DISCUSSION The research results suggest that Galectin-9 and TIM-3, as important immune co-signaling molecules, may play a crucial role in maintaining the immune-tolerant microenvironment at the maternal-fetal interface. Additionally, the association between decidualization and Galectin-9 expression reveals its potential role in pregnancy maintenance, providing new insights for the study of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Jingliang Xu
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China
| | - Xuqing He
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Sujuan Zhang
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China
| | - Li Li
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China.
| | - Penghao Li
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China; Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China; Yunnan Jinxin Jiuzhou Hospital, Yunnan, China.
| |
Collapse
|
8
|
Wang Q, Wang R, Hu H, Huo X, Wang F. Lysosomes' fallback strategies: more than just survival or death. Front Cell Dev Biol 2025; 13:1559504. [PMID: 40134576 PMCID: PMC11933002 DOI: 10.3389/fcell.2025.1559504] [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: 01/12/2025] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Lysosomes are heterogeneous, acidic organelles whose proper functionality is critically dependent on maintaining the integrity of their membranes and the acidity within their lumen. When subjected to stress, the lysosomal membrane can become permeabilized, posing a significant risk to the organelle's survival and necessitating prompt repair. Although numerous mechanisms for lysosomal repair have been identified in recent years, the progression of lysosome-related diseases is more closely linked to the organelle's alternative strategies when repair mechanisms fail, particularly in the contexts of aging and pathogen infection. This review explores lysosomal responses to damage, including the secretion of lysosomal contents and the interactions with lysosome-associated organelles in the endolysosomal system. Furthermore, it examines the role of organelles outside this system, such as the endoplasmic reticulum (ER) and Golgi apparatus, as auxiliary organelles of the endolysosomal system. These alternative strategies are crucial to understanding disease progression. For instance, the secretion and spread of misfolded proteins play key roles in neurodegenerative disease advancement, while pathogen escape via lysosomal secretion and lysosomotropic drug expulsion underlie cancer treatment resistance. Reexamining these lysosomal fallback strategies could provide new perspectives on lysosomal biology and their contribution to disease progression.
Collapse
Affiliation(s)
- Quan Wang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Ruolin Wang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Haihui Hu
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xiaoqing Huo
- Huaian Maternity and Child Healthcare Hospital of JiangSu Province, Huaian, China
| | - Fulong Wang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| |
Collapse
|
9
|
Scott O, Saran E, Freeman SA. The spectrum of lysosomal stress and damage responses: from mechanosensing to inflammation. EMBO Rep 2025; 26:1425-1439. [PMID: 40016424 PMCID: PMC11933331 DOI: 10.1038/s44319-025-00405-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/01/2025] Open
Abstract
Cells and tissues turn over their aged and damaged components in order to adapt to a changing environment and maintain homeostasis. These functions rely on lysosomes, dynamic and heterogeneous organelles that play essential roles in nutrient redistribution, metabolism, signaling, gene regulation, plasma membrane repair, and immunity. Because of metabolic fluctuations and pathogenic threats, lysosomes must adapt in the short and long term to maintain functionality. In response to such challenges, lysosomes deploy a variety of mechanisms that prevent the breaching of their membrane and escape of their contents, including pathogen-associated molecules and hydrolases. While transient permeabilization of the lysosomal membrane can have acute beneficial effects, supporting inflammation and antigen cross-presentation, sustained or repeated lysosomal perforations have adverse metabolic and transcriptional consequences and can lead to cell death. This review outlines factors contributing to lysosomal stress and damage perception, as well as remedial processes aimed at addressing lysosomal disruptions. We conclude that lysosomal stress plays widespread roles in human physiology and pathology, the understanding and manipulation of which can open the door to novel therapeutic strategies.
Collapse
Affiliation(s)
- Ori Scott
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
- Division of Clinical Immunology and Allergy, Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Ekambir Saran
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
10
|
Ma R, Yang W, Guo W, Zhang H, Wang Z, Ge Z. Single-cell transcriptome analysis reveals the dysregulated monocyte state associated with tuberculosis progression. BMC Infect Dis 2025; 25:210. [PMID: 39939918 PMCID: PMC11823163 DOI: 10.1186/s12879-025-10612-3] [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: 06/06/2024] [Accepted: 02/06/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND In tuberculosis (TB) infection, monocytes play a crucial role in regulating the balance between immune tolerance and immune response through various mechanisms. A deeper understanding of the roles of monocyte subsets in TB immune responses may facilitate the development of novel immunotherapeutic strategies and improve TB prevention and treatment. METHODS We retrieved and processed raw single-cell RNA-seq data from SRP247583. Single-cell RNA-seq combined with bioinformatics analysis was employed to investigate the roles of monocytes in TB progression. RESULTS Our findings revealed that classical monocytes expressing inflammatory mediators increased as the disease progressed, whereas non-classical monocytes expressing molecules associated with anti-pathogen infection were progressively depleted. Pseudotime analysis delineated the differentiation trajectory of monocytes from classical to intermediate to non-classical subsets. An abnormal differentiation trajectory to non-classical monocytes may represent a key mechanism underlying TB pathogenesis, with CEBPB and CORO1A identified as genes potentially related to TB development. Analysis of key transcription factors in non-classical monocytes indicated that IRF9 was the only downregulated transcription factor with high AUC activity in this subset. The expression of IRF9 exhibited a decreasing trend in both latent TB infection (LTBI) and active TB groups. Furthermore, dysregulation of transcription factor regulatory networks appeared to impair ferroptosis, with ferroptosis-associated genes MEF2C, MICU1, and PRR5 identified as potential targets of IRF9. Through cell communication analysis, we found that interactions between non-classical monocytes and other subpopulations may mediate TB progression, with MIF and LGALS9 highlighted as potential signaling pathways. CONCLUSION This study employs bioinformatics analysis in conjunction with single-cell sequencing technology to uncover the crucial role of monocyte subsets in tuberculosis infection.
Collapse
Affiliation(s)
- Rong Ma
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wanzhong Yang
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wei Guo
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
| | - Honglai Zhang
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
| | - Zemin Wang
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
| | - Zhaohui Ge
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China.
- General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
11
|
Malik AA, Shariq M, Sheikh JA, Zarin S, Ahuja Y, Fayaz H, Alam A, Ehtesham NZ, Hasnain SE. Activation of the lysosomal damage response and selective autophagy: the coordinated actions of galectins, TRIM proteins, and CGAS-STING1 in providing immunity against Mycobacterium tuberculosis. Crit Rev Microbiol 2025; 51:108-127. [PMID: 38470107 DOI: 10.1080/1040841x.2024.2321494] [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: 08/04/2023] [Revised: 01/16/2024] [Accepted: 02/14/2024] [Indexed: 03/13/2024]
Abstract
Autophagy is a crucial immune defense mechanism that controls the survival and pathogenesis of M. tb by maintaining cell physiology during stress and pathogen attack. The E3-Ub ligases (PRKN, SMURF1, and NEDD4) and autophagy receptors (SQSTM1, TAX1BP1, CALCOCO2, OPTN, and NBR1) play key roles in this process. Galectins (LGALSs), which bind to sugars and are involved in identifying damaged cell membranes caused by intracellular pathogens such as M. tb, are essential. These include LGALS3, LGALS8, and LGALS9, which respond to endomembrane damage and regulate endomembrane damage caused by toxic chemicals, protein aggregates, and intracellular pathogens, including M. tb. They also activate selective autophagy and de novo endolysosome biogenesis. LGALS3, LGALS9, and LGALS8 interact with various components to activate autophagy and repair damage, while CGAS-STING1 plays a critical role in providing immunity against M. tb by activating selective autophagy and producing type I IFNs with antimycobacterial functions. STING1 activates cGAMP-dependent autophagy which provides immunity against various pathogens. Additionally, cytoplasmic surveillance pathways activated by ds-DNA, such as inflammasomes mediated by NLRP3 and AIM2 complexes, control M. tb. Modulation of E3-Ub ligases with small regulatory molecules of LGALSs and TRIM proteins could be a novel host-based therapeutic approach for controlling TB.
Collapse
Affiliation(s)
- Asrar Ahmad Malik
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Mohd Shariq
- ICMR-National Institute of Pathology, New Delhi, India
| | - Javaid Ahmad Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, New Delhi, India
| | - Sheeba Zarin
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, New Delhi, India
| | - Yashika Ahuja
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Haleema Fayaz
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Anwar Alam
- Department of Biotechnology, School of Science and Engineering Technology, Sharda University, Greater Noida, India
| | - Nasreen Z Ehtesham
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Seyed E Hasnain
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| |
Collapse
|
12
|
Ma Y, Zhao C, Feng J, Gou J, Kang E, Guan F, Wu Q, Li X. MSC-sEVs exacerbate senescence by transferring bisecting GlcNAcylated GPNMB. Stem Cell Res Ther 2025; 16:23. [PMID: 39849576 PMCID: PMC11756183 DOI: 10.1186/s13287-025-04140-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND The senescence of bone marrow mesenchymal stem cells (BMMSCs) is increasingly recognized as a critical factor contributing to the pathophysiology of age-related diseases. Recent studies suggest that small extracellular vesicles (sEVs) derived from the serum of elderly individuals may play a pivotal role in promoting BMMSC senescence. Glycoprotein non-metastatic melanoma protein B (GPNMB), a type I transmembrane glycoprotein, is upregulated during cellular senescence and can regulate stem cell ageing. However, the precise mechanisms by which GPNMB influences BMMSCs senescence remain poorly understood. Understanding this relationship could provide valuable insights into therapeutic strategies for enhancing BMMSCs function and mitigating age-related degeneration. METHODS In this study, we conducted comprehensive in vitro experiments to elucidate the effects of sEVs isolated from the serum of elderly donors on the senescence of BMMSCs. We employed advanced proteomic analysis to quantify the expression levels of GPNMB in both BMMSCs and sEVs. Statistical methods were utilized to investigate the correlations between GPNMB expression, glycosylation modifications, and established senescence markers. RESULTS Our findings demonstrate a robust positive correlation between the expression of GPNMB in BMMSCs and sEVs and the induction of cellular senescence. Notably, we observed that elevated levels of GPNMB, particularly those bearing bisecting N-acetylglucosamine (GlcNAc) modifications, significantly enhance the senescent phenotype of BMMSCs. Furthermore, we identified the bisecting GlcNAc modification at the Asn 249 residue of GPNMB as a critical determinant for its senescence-promoting function. CONCLUSIONS This study elucidates the substantial role of sEVs derived from mesenchymal stem cells in exacerbating BMMSC senescence through mechanisms that are critically dependent on the presence of bisecting GlcNAcylated GPNMB. These insights emphasize the necessity of targeting glycosylation modifications of GPNMB in the design of novel senolytic therapies aimed at mitigating cellular ageing and its associated pathologies.
Collapse
Affiliation(s)
- Yihan Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Chongfu Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Jingjing Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Junjie Gou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Enci Kang
- Xi'an Gaoxin No.1 High School International Division, Xi'an, Shaanxi, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Qiong Wu
- The First Affiliated Hospital of Northwest University, Xi'an No.1 Hospital, Xi'an, China.
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Northwest University, Xi'an, China.
| | - Xiang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China.
- Institute of Hematology, Provincial Key Laboratory of Biotechnology, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
13
|
Paddar MA, Wang F, Trosdal ES, Hendrix E, He Y, Salemi MR, Mudd M, Jia J, Duque T, Javed R, Phinney BS, Deretic V. Noncanonical roles of ATG5 and membrane atg8ylation in retromer assembly and function. eLife 2025; 13:RP100928. [PMID: 39773872 PMCID: PMC11706607 DOI: 10.7554/elife.100928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
ATG5 is one of the core autophagy proteins with additional functions such as noncanonical membrane atg8ylation, which among a growing number of biological outputs includes control of tuberculosis in animal models. Here, we show that ATG5 associates with retromer's core components VPS26, VPS29, and VPS35 and modulates retromer function. Knockout of ATG5 blocked trafficking of a key glucose transporter sorted by the retromer, GLUT1, to the plasma membrane. Knockouts of other genes essential for membrane atg8ylation, of which ATG5 is a component, affected GLUT1 sorting, indicating that membrane atg8ylation as a process affects retromer function and endosomal sorting. The contribution of membrane atg8ylation to retromer function in GLUT1 sorting was independent of canonical autophagy. These findings expand the scope of membrane atg8ylation to specific sorting processes in the cell dependent on the retromer and its known interactors.
Collapse
Affiliation(s)
- Masroor Ahmad Paddar
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Fulong Wang
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Einar S Trosdal
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Emily Hendrix
- Department of Chemistry & Chemical Biology, The University of New MexicoAlbuquerqueUnited States
| | - Yi He
- Department of Chemistry & Chemical Biology, The University of New MexicoAlbuquerqueUnited States
| | - Michelle R Salemi
- Proteomics Core Facility, University of California, DavisDavisUnited States
| | - Michal Mudd
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Thabata Duque
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Ruheena Javed
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Brett S Phinney
- Proteomics Core Facility, University of California, DavisDavisUnited States
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of MedicineAlbuquerqueUnited States
- Department of Molecular Genetics and Microbiology, University of New Mexico School of MedicineAlbuquerqueUnited States
| |
Collapse
|
14
|
Zhang CS, Lin SC. Methods to Determine Lysosomal AMPK Activation. Methods Mol Biol 2025; 2882:105-119. [PMID: 39992506 DOI: 10.1007/978-1-0716-4284-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Recent studies have revealed that AMP-activated protein kinase (AMPK) can be activated in a non-canonical, AMP- and ADP-independent manner on the lysosome surface in response to low glucose. This novel mode of activation requires the participation of the glycolytic enzyme aldolase, which acts as a sensor of falling levels of the glucose metabolite fructose-1,6-bisphosphate (FBP). The FBP-unoccupied aldolase blocks the V subfamily of transient receptor potential (TRPV) cation channel to generate a local low calcium environment, under which conditions the inhibited TRPVs contact the lysosome-localized vacuolar ATPase (v-ATPase). This in turn triggers the translocation of AXIN and the associated liver kinase B1 (LKB1) to the lysosomal surface to activate AMPK thereon. Interestingly, such a lysosomal AMPK activating pathway has now been demonstrated to be shared by both the anti-diabetic drug metformin and the anti-tumor/inflammation drug mannose. Here, we describe the experimental conditions and procedures for distinguishing if a certain activation of AMPK is mediated by the lysosomal pathway. We will update the previously described methods for determining AXIN lysosomal translocation and the AMP:ATP and ADP:ATP ratios that are used for differentiating the lysosomal pathway from the canonical, AMP-dependent pathway [1, 2]. We also describe how to determine the lysosomal pH and the inhibition of TRPV activity that are prerequisite for triggering the lysosomal pathway during sensing of low glucose.
Collapse
Affiliation(s)
- Chen-Song Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Sheng-Cai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
15
|
Gu Y, Feng Z, Xu X, Jin L. Identification of a novel immune-related gene signature by single-cell and bulk sequencing for the prediction of the immune landscape and prognosis of breast cancer. Cancer Cell Int 2024; 24:393. [PMID: 39627792 PMCID: PMC11613745 DOI: 10.1186/s12935-024-03589-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND As a common cause of cancer-related deaths in women, BRCA (breast cancer) shows complexity and requires precise biomarkers and treatment methods. This study delves into the molecular makeup of BRCA, focusing on immune profiles, molecular subtypes, gene expression and single-cell analysis. METHODS XCell was used to assess immune infiltration based on TCGA (the Cancer Genome Atlas) data and the clustering analysis was made. Differentially expressed genes were examined in distinct clusters, and the WGCNA (weighted correlation network analysis) was made to establish co-expression networks. The prognostic models were developed by Cox and LASSO-Cox regression. The clustering analysis, GSEA (Gene set enrichment analysis), GSVA (gene set variation analysis) and communication analysis of the single-cell dataset GSE161529 were performed to investigate the functional relevance. Real-time polymerase chain reaction (RT-PCR) was employed for evaluating gene expression. RESULTS The results revealed significant differences in immune cell infiltration between two clusters (C1 and C2). C2 had poorer survival outcomes, which was associated with higher expression of immune checkpoints PD1 and PD-L1. The gene modules identified via WGCNA were correlated with the immune-based subtypes. Then, a prognostic model comprising seven genes (ACSL1, ABCB5, XG, ADH4, OPN4, NPR3, NLGN1) was used to divide patients into high- and low-risk subgroups. The high-risk group had worse prognosis and higher scores of TIDE (Tumor Immune Dysfunction and Exclusion). The single-cell analysis depicted the immune landscape. Macrophages and endothelial cells exhibited higher AUCell scores. In cellular communication analysis, notably significant ligand-receptor interactions of HLA-DRA-> CD4 and TNFSF13B-> HLA-DPB1 were observed. The proportion of endothelial cells was correlated with risk scores. Finally, RT-PCR results illustrated the expression of seven genes in BRCA specimens. CONCLUSION The integrative analysis provides new insights into molecular complexities of BRCA. Immune profiles and gene signatures hold potential for improving stratification of BRCA patients and guiding the development of personalized immunotherapy strategies.
Collapse
Affiliation(s)
- Yanlin Gu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Zhengyang Feng
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Xiaoyan Xu
- Department of Operating Room, Traditional Chinese Medicine Hospital of Kunshan, Jiangsu, China
| | - Liyan Jin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu, China.
| |
Collapse
|
16
|
Gu H, Qiu H, Yang H, Deng Z, Zhang S, Du L, He F. PRRSV utilizes MALT1-regulated autophagy flux to switch virus spread and reserve. Autophagy 2024; 20:2697-2718. [PMID: 39081059 PMCID: PMC11587858 DOI: 10.1080/15548627.2024.2386195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/03/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a major swine pathogen, which can survive host antiviral immunity with various mechanisms. PRRSV infection induces macroautophagy/autophagy, facilitating virus replication. MALT1, a central immune regulator, was manipulated by PRRSV to optimize viral infection at different stages of the virus cycle. In this study, the key role of MALT1 in autophagy regulation during PRRSV infection was characterized, enlightening the role of autophagy flux in favor of virus spread and persistent infection. PRRSV-induced autophagy was confirmed to facilitate virus proliferation. Furthermore, autophagic fusion was dynamically regulated during PRRSV infection. Importantly, PRRSV-induced MALT1 facilitated autophagosome-lysosome fusion and autolysosome formation, thus contributing to autophagy flux and virus proliferation. Mechanically, MALT1 regulated autophagy via mediating MTOR-ULK1 and -TFEB signaling and affecting lysosomal homeostasis. MALT1 inhibition by inhibitor Mi-2 or RNAi induced lysosomal membrane permeabilization (LMP), leading to the block of autophagic fusion. Further, MALT1 overexpression alleviated PRRSV-induced LMP via inhibiting ROS generation. In addition, blocking autophagy flux suppressed virus release significantly, indicating that MALT1-maintained complete autophagy flux during PRRSV infection favors successful virus spread and its proliferation. In contrast, autophagosome accumulation upon MALT1 inhibition promoted PRRSV reserve for future virus proliferation once the autophagy flux recovers. Taken together, for the first time, these findings elucidate that MALT1 was utilized by PRRSV to regulate host autophagy flux, to determine the fate of virus for either proliferation or reserve.Abbreviations: 3-MA: 3-methyladenine; BafA1: bafilomycin A1; BFP/mBFP: monomeric blue fluorescent protein; CQ: chloroquine; DMSO: dimethyl sulfoxide; dsRNA: double-stranded RNA; GFP: green fluorescent protein; hpi: hours post infection; IFA: indirect immunofluorescence assay; LAMP1: lysosomal associated membrane protein 1; LGALS3: galectin 3; LLOMe: L-leucyl-L-leucine-methyl ester; LMP: lysosomal membrane permeabilization; mAb: monoclonal antibody; MALT1: MALT1 paracaspase; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MOI: multiplicity of infection; MTOR: mechanistic target of rapamycin kinase; NFKB/NF-κB: nuclear factor kappa B; nsp: nonstructural protein; ORF: open reading frame; pAb: polyclonal antibody; PRRSV: porcine reproductive and respiratory syndrome virus; PRRSV-N: PRRSV nucleocapsid protein; Rapa: rapamycin; RFP: red fluorescent protein; ROS: reactive oxygen species; SBI: SBI-0206965; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; TCID50: 50% tissue culture infective dose; TFEB: transcription factor EB; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Han Gu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
- TianMu Laboratory, ZJU-Xinchang Joint Innovation Centre, Xinchang, Zhejiang, P.R. China
| | - He Qiu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
- TianMu Laboratory, ZJU-Xinchang Joint Innovation Centre, Xinchang, Zhejiang, P.R. China
| | - Haotian Yang
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
- TianMu Laboratory, ZJU-Xinchang Joint Innovation Centre, Xinchang, Zhejiang, P.R. China
| | - Zhuofan Deng
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
- TianMu Laboratory, ZJU-Xinchang Joint Innovation Centre, Xinchang, Zhejiang, P.R. China
| | - Shengkun Zhang
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
- TianMu Laboratory, ZJU-Xinchang Joint Innovation Centre, Xinchang, Zhejiang, P.R. China
| | - Liuyang Du
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Fang He
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
- TianMu Laboratory, ZJU-Xinchang Joint Innovation Centre, Xinchang, Zhejiang, P.R. China
| |
Collapse
|
17
|
Currais A, Kepchia D, Liang Z, Maher P. The Role of AMP-activated Protein Kinase in Oxytosis/Ferroptosis: Protector or Potentiator? Antioxid Redox Signal 2024; 41:e1173-e1186. [PMID: 35243895 PMCID: PMC11693968 DOI: 10.1089/ars.2022.0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/20/2023]
Abstract
Significance: Evidence for a role for the oxytosis/ferroptosis regulated cell death pathway in aging and neurodegenerative diseases has been growing over the past few years. Because of this, there is an increasing necessity to identify endogenous signaling pathways that can be modulated to protect cells from this form of cell death. Recent Advances: Recently, several studies have identified a protective role for the AMP-activated protein kinase (AMPK)/acetyl CoA carboxylase 1 (ACC1) pathway in oxytosis/ferroptosis. However, there are also a number of studies suggesting that this pathway contributes to cell death initiated by various inducers of oxytosis/ferroptosis. Critical Issues: The goals of this review are to provide an overview and analysis of the published studies and highlight specific areas where more research is needed. Future Directions: Much remains to be learned about AMPK signaling in oxytosis/ferroptosis, especially the conditions where it is protective. Furthermore, the role of AMPK signaling in the brain and especially the aging brain needs further investigation.
Collapse
Affiliation(s)
- Antonio Currais
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Devin Kepchia
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Zhibin Liang
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| |
Collapse
|
18
|
Duran J, Salinas JE, Wheaton RP, Poolsup S, Allers L, Rosas-Lemus M, Chen L, Cheng Q, Pu J, Salemi M, Phinney B, Ivanov P, Lystad AH, Bhaskar K, Rajaiya J, Perkins DJ, Jia J. Calcium signaling from damaged lysosomes induces cytoprotective stress granules. EMBO J 2024; 43:6410-6443. [PMID: 39533058 PMCID: PMC11649789 DOI: 10.1038/s44318-024-00292-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/18/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Lysosomal damage induces stress granule (SG) formation. However, the importance of SGs in determining cell fate and the precise mechanisms that mediate SG formation in response to lysosomal damage remain unclear. Here, we describe a novel calcium-dependent pathway controlling SG formation, which promotes cell survival during lysosomal damage. Mechanistically, the calcium-activated protein ALIX transduces lysosomal damage signals to SG formation by controlling eIF2α phosphorylation after sensing calcium leakage. ALIX enhances eIF2α phosphorylation by promoting the association between PKR and its activator PACT, with galectin-3 inhibiting this interaction; these regulatory events occur on damaged lysosomes. We further find that SG formation plays a crucial role in promoting cell survival upon lysosomal damage caused by factors such as SARS-CoV-2ORF3a, adenovirus, malarial pigment, proteopathic tau, or environmental hazards. Collectively, these data provide insights into the mechanism of SG formation upon lysosomal damage and implicate it in diseases associated with damaged lysosomes and SGs.
Collapse
Affiliation(s)
- Jacob Duran
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Jay E Salinas
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Rui Ping Wheaton
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Suttinee Poolsup
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Monica Rosas-Lemus
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Li Chen
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Qiuying Cheng
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Jing Pu
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Michelle Salemi
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, CA, 95616, USA
| | - Brett Phinney
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, CA, 95616, USA
| | - Pavel Ivanov
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; HMS Initiative for RNA Medicine, Boston, MA, 02115, USA
| | - Alf Håkon Lystad
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Jaya Rajaiya
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Douglas J Perkins
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Jingyue Jia
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA.
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA.
| |
Collapse
|
19
|
Qin L, Xu J, Chen J, Wang S, Zheng R, Cui Z, Liu Z, Wu X, Wang J, Huang X, Wang Z, Wang M, Pan R, Kaufmann SHE, Meng X, Zhang L, Sha W, Liu H. Cell-autonomous targeting of arabinogalactan by host immune factors inhibits mycobacterial growth. eLife 2024; 13:RP92737. [PMID: 39495223 PMCID: PMC11534329 DOI: 10.7554/elife.92737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
Deeper understanding of the crosstalk between host cells and Mycobacterium tuberculosis (Mtb) provides crucial guidelines for the rational design of novel intervention strategies against tuberculosis (TB). Mycobacteria possess a unique complex cell wall with arabinogalactan (AG) as a critical component. AG has been identified as a virulence factor of Mtb which is recognized by host galectin-9. Here, we demonstrate that galectin-9 directly inhibited mycobacterial growth through AG-binding property of carbohydrate-recognition domain 2. Furthermore, IgG antibodies with AG specificity were detected in the serum of TB patients. Based on the interaction between galectin-9 and AG, we developed a monoclonal antibody (mAb) screening assay and identified AG-specific mAbs which profoundly inhibit Mtb growth. Mechanistically, proteomic profiling and morphological characterizations revealed that AG-specific mAbs regulate AG biosynthesis, thereby inducing cell wall swelling. Thus, direct AG-binding by galectin-9 or antibodies contributes to protection against TB. Our findings pave the way for the rational design of novel immunotherapeutic strategies for TB control.
Collapse
Affiliation(s)
- Lianhua Qin
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Junfang Xu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Sen Wang
- Department of Infectious Diseases, National Medical Centre for Infectious Diseases, National Clinical Research Centre for Aging and Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Ruijuan Zheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Zhenling Cui
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Zhonghua Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiangyang Wu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Jie Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaochen Huang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | | | | | | | - Stefan HE Kaufmann
- Max Planck Institute for Infection BiologyBerlinGermany
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Hagler Institute for Advanced Study, Texas A&M UniversityCollege StationUnited States
| | - Xun Meng
- Abmart IncShanghaiChina
- Multitude TherapeuticsShanghaiChina
| | - Lu Zhang
- School of Life Science, Fudan UniversityShanghaiChina
| | - Wei Sha
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Haipeng Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| |
Collapse
|
20
|
Paddar MA, Wang F, Trosdal ES, Hendrix E, He Y, Salemi M, Mudd M, Jia J, Duque TLA, Javed R, Phinney B, Deretic V. Noncanonical roles of ATG5 and membrane atg8ylation in retromer assembly and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602886. [PMID: 39026874 PMCID: PMC11257513 DOI: 10.1101/2024.07.10.602886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
ATG5 is one of the core autophagy proteins with additional functions such as noncanonical membrane atg8ylation, which among a growing number of biological outputs includes control of tuberculosis in animal models. Here we show that ATG5 associates with retromer's core components VPS26, VPS29 and VPS35 and modulates retromer function. Knockout of ATG5 blocked trafficking of a key glucose transporter sorted by the retromer, GLUT1, to the plasma membrane. Knockouts of other genes essential for membrane atg8ylation, of which ATG5 is a component, affected GLUT1 sorting, indicating that membrane atg8ylation as a process affects retromer function and endosomal sorting. The contribution of membrane atg8ylation to retromer function in GLUT1 sorting was independent of canonical autophagy. These findings expand the scope of membrane atg8ylation to specific sorting processes in the cell dependent on the retromer and its known interactors.
Collapse
Affiliation(s)
- Masroor Ahmad Paddar
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Fulong Wang
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Einar S Trosdal
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Emily Hendrix
- Department of Chemistry & Chemical Biology, The University of New Mexico, Albuquerque, NM, USA
| | - Yi He
- Department of Chemistry & Chemical Biology, The University of New Mexico, Albuquerque, NM, USA
| | - Michelle Salemi
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, CA 95616, USA
| | - Michal Mudd
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Thabata L A Duque
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ruheena Javed
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Brett Phinney
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, CA 95616, USA
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
- Lead Contact
| |
Collapse
|
21
|
Salminen A. Inhibitory immune checkpoints suppress the surveillance of senescent cells promoting their accumulation with aging and in age-related diseases. Biogerontology 2024; 25:749-773. [PMID: 38954358 PMCID: PMC11374851 DOI: 10.1007/s10522-024-10114-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The accumulation of pro-inflammatory senescent cells within tissues is a common hallmark of the aging process and many age-related diseases. This modification has been called the senescence-associated secretory phenotype (SASP) and observed in cultured cells and in cells isolated from aged tissues. Currently, there is a debate whether the accumulation of senescent cells within tissues should be attributed to increased generation of senescent cells or to a defect in their elimination from aging tissues. Emerging studies have revealed that senescent cells display an increased expression of several inhibitory immune checkpoint ligands, especially those of the programmed cell death protein-1 (PD-1) ligand-1 (PD-L1) proteins. It is known that the PD-L1 ligands, especially those of cancer cells, target the PD-1 receptor of cytotoxic CD8+ T and natural killer (NK) cells disturbing their functions, e.g., evoking a decline in their cytotoxic activity and promoting their exhaustion and even apoptosis. An increase in the level of the PD-L1 protein in senescent cells was able to suppress their immune surveillance and inhibit their elimination by cytotoxic CD8+ T and NK cells. Senescent cells are known to express ligands for several inhibitory immune checkpoint receptors, i.e., PD-1, LILRB4, NKG2A, TIM-3, and SIRPα receptors. Here, I will briefly describe those pathways and examine whether these inhibitory checkpoints could be involved in the immune evasion of senescent cells with aging and age-related diseases. It seems plausible that an enhanced inhibitory checkpoint signaling can prevent the elimination of senescent cells from tissues and thus promote the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
22
|
Ferrari V, Tedesco B, Cozzi M, Chierichetti M, Casarotto E, Pramaggiore P, Cornaggia L, Mohamed A, Patelli G, Piccolella M, Cristofani R, Crippa V, Galbiati M, Poletti A, Rusmini P. Lysosome quality control in health and neurodegenerative diseases. Cell Mol Biol Lett 2024; 29:116. [PMID: 39237893 PMCID: PMC11378602 DOI: 10.1186/s11658-024-00633-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Lysosomes are acidic organelles involved in crucial intracellular functions, including the degradation of organelles and protein, membrane repair, phagocytosis, endocytosis, and nutrient sensing. Given these key roles of lysosomes, maintaining their homeostasis is essential for cell viability. Thus, to preserve lysosome integrity and functionality, cells have developed a complex intracellular system, called lysosome quality control (LQC). Several stressors may affect the integrity of lysosomes, causing Lysosomal membrane permeabilization (LMP), in which membrane rupture results in the leakage of luminal hydrolase enzymes into the cytosol. After sensing the damage, LQC either activates lysosome repair, or induces the degradation of the ruptured lysosomes through autophagy. In addition, LQC stimulates the de novo biogenesis of functional lysosomes and lysosome exocytosis. Alterations in LQC give rise to deleterious consequences for cellular homeostasis. Specifically, the persistence of impaired lysosomes or the malfunctioning of lysosomal processes leads to cellular toxicity and death, thereby contributing to the pathogenesis of different disorders, including neurodegenerative diseases (NDs). Recently, several pieces of evidence have underlined the importance of the role of lysosomes in NDs. In this review, we describe the elements of the LQC system, how they cooperate to maintain lysosome homeostasis, and their implication in the pathogenesis of different NDs.
Collapse
Affiliation(s)
- Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Paola Pramaggiore
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Laura Cornaggia
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Ali Mohamed
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Guglielmo Patelli
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy.
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| |
Collapse
|
23
|
Wang ML, Zhang YJ, He DL, Li T, Zhao MM, Zhao LM. Inhibition of PLA2G4A attenuated valproic acid- induced lysosomal membrane permeabilization and restored impaired autophagic flux: Implications for hepatotoxicity. Biochem Pharmacol 2024; 227:116438. [PMID: 39025409 DOI: 10.1016/j.bcp.2024.116438] [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: 02/18/2024] [Revised: 07/05/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Valproic acid (VPA) has broad efficacy against several seizures but causes liver injury limiting its prolonged clinical use. Some studies have demonstrated that VPA-induced hepatotoxicity is characterized by microvesicular hepatic steatosis. However, novel detailed mechanisms to explain VPA-induced hepatic steatosis and experimentally rigorously validated protective agents are still lacking. In this study, 8-week-old C57BL/6J mice were gavaged with VPA (500 mg/kg/d) for 4 weeks to establish an in vivo model of VPA-induced chronic liver injury. Quantitative proteomic and non-targeted lipidomic analyses were performed to explore the underlying mechanisms of VPA-induced hepatotoxicity. As a result, VPA-induced hepatotoxicity is associated with impaired autophagic flux, which is attributed to lysosomal dysfunction. Further studies revealed that VPA-induced lysosomal membrane permeabilization (LMP), allows soluble lysosomal enzymes to leak into the cytosol, which subsequently led to impaired lysosomal acidification. A lower abundance of glycerophospholipids and an increased abundance of lysophospholipids in liver tissues of mice in the VPA group strongly indicated that VPA-induced LMP may be mediated by the activation of phospholipase PLA2G4A. Metformin (Met) acted as a potential protective agent attenuating VPA-induced liver dysfunction and excessive lipid accumulation. Molecular docking and cellular thermal shift assays demonstrated that Met inhibited the activity of PLA2G4A by directly binding to it, thereby ameliorating VPA-induced LMP and autophagic flux impairment. In conclusion, this study highlights the therapeutic potential of targeting PLA2G4A-mediated lysosomal dysfunction in VPA-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ming-Lu Wang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu-Jia Zhang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Da-Long He
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, Liaoning, China
| | - Tong Li
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ming-Ming Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Li-Mei Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
24
|
Domingues N, Catarino S, Cristóvão B, Rodrigues L, Carvalho FA, Sarmento MJ, Zuzarte M, Almeida J, Ribeiro-Rodrigues T, Correia-Rodrigues Â, Fernandes F, Rodrigues-Santos P, Aasen T, Santos NC, Korolchuk VI, Gonçalves T, Milosevic I, Raimundo N, Girão H. Connexin43 promotes exocytosis of damaged lysosomes through actin remodelling. EMBO J 2024; 43:3627-3649. [PMID: 39044100 PMCID: PMC11377567 DOI: 10.1038/s44318-024-00177-3] [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: 12/11/2023] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
A robust and efficient cellular response to lysosomal membrane damage prevents leakage from the lysosome lumen into the cytoplasm. This response is understood to happen through either lysosomal membrane repair or lysophagy. Here we report exocytosis as a third response mechanism to lysosomal damage, which is further potentiated when membrane repair or lysosomal degradation mechanisms are impaired. We show that Connexin43 (Cx43), a protein canonically associated with gap junctions, is recruited from the plasma membrane to damaged lysosomes, promoting their secretion and accelerating cell recovery. The effects of Cx43 on lysosome exocytosis are mediated by a reorganization of the actin cytoskeleton that increases plasma membrane fluidity and decreases cell stiffness. Furthermore, we demonstrate that Cx43 interacts with the actin nucleator Arp2, the activity of which was shown to be necessary for Cx43-mediated actin rearrangement and lysosomal exocytosis following damage. These results define a novel mechanism of lysosomal quality control whereby Cx43-mediated actin remodelling potentiates the secretion of damaged lysosomes.
Collapse
Affiliation(s)
- Neuza Domingues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Steve Catarino
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Beatriz Cristóvão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria João Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Jani Almeida
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Teresa Ribeiro-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Ânia Correia-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Fábio Fernandes
- Institute for Bioengineering and Biosciences (IBB) and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo Rodrigues-Santos
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Trond Aasen
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Teresa Gonçalves
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- University of Oxford, Centre for Human Genetics, Nuffield Department of Medicine, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Henrique Girão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal.
| |
Collapse
|
25
|
Bonet-Ponce L, Kluss JH, Cookson MR. Mechanisms of lysosomal tubulation and sorting driven by LRRK2. Biochem Soc Trans 2024; 52:1909-1919. [PMID: 39083004 PMCID: PMC11668303 DOI: 10.1042/bst20240087] [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: 05/14/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024]
Abstract
Lysosomes are dynamic cellular structures that adaptively remodel their membrane in response to stimuli, including membrane damage. Lysosomal dysfunction plays a central role in the pathobiology of Parkinson's disease (PD). Gain-of-function mutations in Leucine-rich repeat kinase 2 (LRRK2) cause familial PD and genetic variations in its locus increase the risk of developing the sporadic form of the disease. We previously uncovered a process we term LYTL (LYsosomal Tubulation/sorting driven by LRRK2), wherein membrane-damaged lysosomes generate tubules sorted into mobile vesicles. Subsequently, these vesicles interact with healthy lysosomes. LYTL is orchestrated by LRRK2 kinase activity, via the recruitment and phosphorylation of a subset of RAB GTPases. Here, we summarize the current understanding of LYTL and its regulation, as well as the unknown aspects of this process.
Collapse
Affiliation(s)
- Luis Bonet-Ponce
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, U.S.A
| | | | - Mark R. Cookson
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, U.S.A
| |
Collapse
|
26
|
Deretic V, Duque T, Trosdal E, Paddar M, Javed R, Akepati P. Membrane atg8ylation in Canonical and Noncanonical Autophagy. J Mol Biol 2024; 436:168532. [PMID: 38479594 PMCID: PMC11260254 DOI: 10.1016/j.jmb.2024.168532] [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: 01/23/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024]
Abstract
Membrane atg8ylation is a homeostatic process responding to membrane remodeling and stress signals. Membranes are atg8ylated by mammalian ATG8 ubiquitin-like proteins through a ubiquitylation-like cascade. A model has recently been put forward which posits that atg8ylation of membranes is conceptually equivalent to ubiquitylation of proteins. Like ubiquitylation, membrane atg8ylation involves E1, E2 and E3 enzymes. The E3 ligases catalyze the final step of atg8ylation of aminophospholipids in membranes. Until recently, the only known E3 ligase for membrane atg8ylation was ATG16L1 in a noncovalent complex with the ATG12-ATG5 conjugate. ATG16L1 was first identified as a factor in canonical autophagy. During canonical autophagy, the ATG16L1-based E3 ligase complex includes WIPI2, which in turn recognizes phosphatidylinositiol 3-phosphate and directs atg8ylation of autophagic phagophores. As an alternative to WIPIs, binding of ATG16L1 to the proton pump V-ATPase guides atg8ylation of endolysosomal and phagosomal membranes in response to lumenal pH changes. Recently, a new E3 complex containing TECPR1 instead of ATG16L1, has been identified that responds to sphingomyelin's presence on the cytofacial side of perturbed endolysosomal membranes. In present review, we cover the principles of membrane atg8ylation, catalog its various presentations, and provide a perspective on the growing repertoire of E3 ligase complexes directing membrane atg8ylation at diverse locations.
Collapse
Affiliation(s)
- Vojo Deretic
- Autophagy Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA.
| | - Thabata Duque
- Autophagy Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Einar Trosdal
- Autophagy Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Masroor Paddar
- Autophagy Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ruheena Javed
- Autophagy Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Prithvi Akepati
- Gastroenterology Division, Department of Internal Medicine, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| |
Collapse
|
27
|
Zhang C, Liu X, Liu X, Hua R, Liu H, Ma J, Zou D, Wang G, Yuan Q, Wang B, Wei S, Chen Y. Adenosine kinase protects against acetaminophen-induced acute liver injury by activating autophagy in hepatocytes. Cell Biol Toxicol 2024; 40:59. [PMID: 39060559 PMCID: PMC11281981 DOI: 10.1007/s10565-024-09906-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Acute liver injury (ALI) is a common life-threatening condition with a high mortality rate due to liver disease-related death. However, current therapeutic interventions for ALI remain ineffective, and the development of effective novel therapies is urgently needed. Liver samples from patients with drug-induced ALI were collected to detect adenosine kinase (ADK) expression. Male C57BL/6 J mice, hepatocyte-specific ADK knockout (ADKHKO) mice, and their controls (ADKf/f) were exposed to acetaminophen (APAP) and other treatments to investigate the mechanisms of APAP-related ALI. ADK expression was significantly decreased in APAP-injured livers. Hepatocyte-specific ADK deficiency exacerbated APAP-induced ALI, while a gain-of-function approach delivering AAV-ADK, markedly alleviated APAP-induced ALI, as indicated by changes in alanine aminotransferases (ALT) levels, aspartate aminotransferase (AST) levels, neutrophil infiltration and hepatocyte death. This study showed that ADK played a critical role in ALI by activating autophagy through two signaling pathways, the adenosine monophosphate-activated protein kinase (AMPK)-mTOR pathway and the adenosine receptor A1 (ADORA1)-Akt-mTOR pathway. Furthermore, we found that metformin upregulated ADK expression in hepatocytes and protected against APAP-induced ALI. These results demonstrate that ADK is critical in protecting against APAP-induced ALI and that developing therapeutics targeting ADK-adenosine-ADORA1 is a new approach for ALI treatment. Metformin is a potential candidate for preventing ALI by upregulating ADK.
Collapse
Affiliation(s)
- Chuanxin Zhang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xilong Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Rui Hua
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Han Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jiaxin Ma
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Dan Zou
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Guangmei Wang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Qiuhuan Yuan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Bailu Wang
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
28
|
Lo TH, Weng IC, Chen HL, Liu FT. The role of galectins in the regulation of autophagy and inflammasome in host immunity. Semin Immunopathol 2024; 46:6. [PMID: 39042263 DOI: 10.1007/s00281-024-01018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024]
Abstract
Galectins, a family of glycan-binding proteins have been shown to bind a wide range of glycans. In the cytoplasm, these glycans can be endogenous (or "self"), originating from damaged endocytic vesicles, or exogenous (or "non-self"), found on the surface of invading microbial pathogens. Galectins can detect these unusual cytosolic exposures to glycans and serve as critical regulators in orchestrating immune responses in innate and adaptive immunity. This review provides an overview of how galectins modulate host cellular responses, such as autophagy, xenophagy, and inflammasome-dependent cell death program, to infection.
Collapse
Affiliation(s)
- Tzu-Han Lo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - I-Chun Weng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Hung-Lin Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
- Department of Dermatology, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
29
|
Jacob R, Gorek LS. Intracellular galectin interactions in health and disease. Semin Immunopathol 2024; 46:4. [PMID: 38990375 PMCID: PMC11239732 DOI: 10.1007/s00281-024-01010-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/07/2024] [Indexed: 07/12/2024]
Abstract
In the galectin family, a group of lectins is united by their evolutionarily conserved carbohydrate recognition domains. These polypeptides play a role in various cellular processes and are implicated in disease mechanisms such as cancer, fibrosis, infection, and inflammation. Following synthesis in the cytosol, manifold interactions of galectins have been described both extracellularly and intracellularly. Extracellular galectins frequently engage with glycoproteins or glycolipids in a carbohydrate-dependent manner. Intracellularly, galectins bind to non-glycosylated proteins situated in distinct cellular compartments, each with multiple cellular functions. This diversity complicates attempts to form a comprehensive understanding of the role of galectin molecules within the cell. This review enumerates intracellular galectin interaction partners and outlines their involvement in cellular processes. The intricate connections between galectin functions and pathomechanisms are illustrated through discussions of intracellular galectin assemblies in immune and cancer cells. This underscores the imperative need to fully comprehend the interplay of galectins with the cellular machinery and to devise therapeutic strategies aimed at counteracting the establishment of galectin-based disease mechanisms.
Collapse
Affiliation(s)
- Ralf Jacob
- Department of Cell Biology and Cell Pathology, Philipps University of Marburg, Karl-von-Frisch-Str. 14, D-35043, Marburg, Germany.
| | - Lena-Sophie Gorek
- Department of Cell Biology and Cell Pathology, Philipps University of Marburg, Karl-von-Frisch-Str. 14, D-35043, Marburg, Germany
| |
Collapse
|
30
|
Cheng Y, Wang S, Gao Q, Fang D. ATXN3 functions as a tumor suppressor through potentiating galectin-9-mediated apoptosis in human colon adenocarcinoma. J Biol Chem 2024; 300:107415. [PMID: 38815863 PMCID: PMC11254720 DOI: 10.1016/j.jbc.2024.107415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024] Open
Abstract
While deubiquitinase ATXN3 has been implicated as a potential oncogene in various types of human cancers, its role in colon adenocarcinoma remains understudied. Surprisingly, our findings demonstrate that ATXN3 exerts an antitumor effect in human colon cancers through potentiating Galectin-9-induced apoptosis. CRISPR-mediated ATXN3 deletion unexpectedly intensified colon cancer growth both in vitro and in xenograft colon cancers. At the molecular level, we identified ATXN3 as a bona fide deubiquitinase specifically targeting Galectin-9, as ATXN3 interacted with and inhibited Galectin-9 ubiquitination. Consequently, targeted ATXN3 ablation resulted in reduced Galectin-9 protein expression, thereby diminishing Galectin-9-induced colon cancer apoptosis and cell growth arrest. The ectopic expression of Galectin-9 fully reversed the growth of ATXN3-null colon cancer in mice. Furthermore, immunohistochemistry staining revealed a significant reduction in both ATXN3 and Galectin-9 protein expression, along with a positive correlation between them in human colon cancer. Our study identifies the first Galectin-9-specific deubiquitinase and unveils a tumor-suppressive role of ATXN3 in human colon cancer.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Shengnan Wang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Qiong Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Center for Human Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
31
|
Cui Y, Liu Q, Zhang Q, Di X, Zhang H. Benzoylaconine Protects Skeletal Muscle Against Ischemia-Reperfusion Injury Through Activation of IF1-Dependent AMPK/Nrf2 Axis. Drug Des Devel Ther 2024; 18:2125-2142. [PMID: 38882050 PMCID: PMC11178076 DOI: 10.2147/dddt.s456699] [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: 12/26/2023] [Accepted: 06/07/2024] [Indexed: 06/18/2024] Open
Abstract
Background Aconitum carmichaelii (Fuzi) has been conventionally used to cure a variety of ailments, such as pain, cold sensations, and numbness of limb muscles (Bi Zheng) in China. Our prior investigations identified Benzoylaconine (BAC) as a bioactive alkaloid derived from Aconitum carmichaelii, with other studies also demonstrating its significant pharmacological potential. Purpose This study aimed to explore the potential of BAC as a protective agent against skeletal muscle ischemia-reperfusion (I/R) injury and to elucidate the underlying mechanisms. Methods In vivo models involved subjecting Sprague-Dawley rats to I/R through femoral artery ligation followed by reperfusion, while in vitro models utilized C2C12 cells subjected to hypoxia/reoxygenation (H/R). CCK-8 assay was used to assess cell viability. TUNEL staining and flow cytometric analysis were used to measure cell apoptosis. Biochemical assay was used to assess skeletal muscle injury and oxidative stress. Immunofluorescence and Western blot were performed to determine protein levels. Results BAC effectively protected muscle tissue from I/R injury, enhancing cell viability (p<0.01), elevating SOD levels (p<0.05), and reducing CK (p<0.01), LDH (p<0.01), ROS (p<0.01), MDA (p<0.01), and apoptosis-related molecules in vivo and in vitro (p<0.05, p<0.01). Mechanistically, BAC increased the expression of IF1, phosphorylated AMPK, facilitated the translocation of nuclear Nrf2, and induced the expression of HO-1 (p<0.01). Notably, AMPK inhibitor Compound C significantly hindered the ability of BAC to ameliorate H/R-induced cell injury (p<0.05), oxidative stress(p<0.01), and apoptosis (p<0.05), as well as promote Nrf2 nuclear translocation (p<0.01). Moreover, silencing of IF1 with siRNA abolished BAC-induced activation of AMPK/Nrf2 axis (p<0.01). Conclusion Our study provides novel evidence supporting the potential of BAC as a myocyte-protective agent against I/R injury, and we establish a previously unknown mechanism involving the activation of the IF1-dependent AMPK/Nrf2 axis in mediating the protective effects of BAC.
Collapse
Affiliation(s)
- Yidong Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People’s Republic of China
| | - Qingming Liu
- Department of Neurology, Shandong Second Provincial General Hospital, Jinan, 250012, People’s Republic of China
| | - Qiqiang Zhang
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, People’s Republic of China
| | - Xuemei Di
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, People’s Republic of China
| | - Hai Zhang
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, People’s Republic of China
| |
Collapse
|
32
|
Chen P, Cao XW, Dong JW, Zhao J, Wang FJ. Saponin and Ribosome-Inactivating Protein Synergistically Trigger Lysosome-Dependent Apoptosis by Inhibiting Lysophagy: Potential to Become a New Antitumor Strategy. Mol Pharm 2024; 21:2993-3005. [PMID: 38722865 DOI: 10.1021/acs.molpharmaceut.4c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The susceptibility of lysosomal membranes in tumor cells to cationic amphiphilic drugs (CADs) enables CADs to induce lysosomal membrane permeabilization (LMP) and trigger lysosome-dependent cell death (LDCD), suggesting a potential antitumor therapeutic approach. However, the existence of intrinsic lysosomal damage response mechanisms limits the display of the pharmacological activity of CADs. In this study, we report that low concentrations of QS-21, a saponin with cationic amphiphilicity extracted from Quillaja Saponaria tree, can induce LMP but has nontoxicity to tumor cells. QS-21 and MAP30, a type I ribosome-inactivating protein, synergistically induce apoptosis in tumor cells at low concentrations of both. Mechanistically, QS-21-induced LMP helps MAP30 escape from endosomes or lysosomes and subsequently enter the endoplasmic reticulum, where MAP30 downregulates the expression of autophagy-associated LC3 proteins, thereby inhibiting lysophagy. The inhibition of lysophagy results in the impaired clearance of damaged lysosomes, leading to the leakage of massive lysosomal contents such as cathepsins into the cytoplasm, ultimately triggering LDCD. In summary, our study showed that coadministration of QS-21 and MAP30 amplified the lysosomal disruption and can be a new synergistic LDCD-based antitumor therapy.
Collapse
Affiliation(s)
- Piao Chen
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Xue-Wei Cao
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
| | - Jing-Wen Dong
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
| | - Jian Zhao
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Fu-Jun Wang
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| |
Collapse
|
33
|
Duran J, Poolsup S, Allers L, Lemus MR, Cheng Q, Pu J, Salemi M, Phinney B, Jia J. A mechanism that transduces lysosomal damage signals to stress granule formation for cell survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587368. [PMID: 38617306 PMCID: PMC11014484 DOI: 10.1101/2024.03.29.587368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Lysosomal damage poses a significant threat to cell survival. Our previous work has reported that lysosomal damage induces stress granule (SG) formation. However, the importance of SG formation in determining cell fate and the precise mechanisms through which lysosomal damage triggers SG formation remains unclear. Here, we show that SG formation is initiated via a novel calcium-dependent pathway and plays a protective role in promoting cell survival in response to lysosomal damage. Mechanistically, we demonstrate that during lysosomal damage, ALIX, a calcium-activated protein, transduces lysosomal damage signals by sensing calcium leakage to induce SG formation by controlling the phosphorylation of eIF2α. ALIX modulates eIF2α phosphorylation by regulating the association between PKR and its activator PACT, with galectin-3 exerting a negative effect on this process. We also found this regulatory event of SG formation occur on damaged lysosomes. Collectively, these investigations reveal novel insights into the precise regulation of SG formation triggered by lysosomal damage, and shed light on the interaction between damaged lysosomes and SGs. Importantly, SG formation is significant for promoting cell survival in the physiological context of lysosomal damage inflicted by SARS-CoV-2 ORF3a, adenovirus infection, Malaria hemozoin, proteopathic tau as well as environmental hazard silica.
Collapse
Affiliation(s)
- Jacob Duran
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM 87106, USA
| | - Suttinee Poolsup
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM 87106, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA
| | - Monica Rosas Lemus
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA
| | - Qiuying Cheng
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA
| | - Jing Pu
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA
| | - Michelle Salemi
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, CA 95616, USA
| | - Brett Phinney
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, CA 95616, USA
| | - Jingyue Jia
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM 87106, USA
- Lead Contact
| |
Collapse
|
34
|
Peltan EL, Riley NM, Flynn RA, Roberts DS, Bertozzi CR. Galectin-3 does not interact with RNA directly. Glycobiology 2024; 34:cwad076. [PMID: 37815932 PMCID: PMC11648975 DOI: 10.1093/glycob/cwad076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 10/12/2023] Open
Abstract
Galectin-3, well characterized as a glycan binding protein, has been identified as a putative RNA binding protein, possibly through participation in pre-mRNA maturation through interactions with splicosomes. Given recent developments with cell surface RNA biology, the putative dual-function nature of galectin-3 evokes a possible non-classical connection between glycobiology and RNA biology. However, with limited functional evidence of a direct RNA interaction, many molecular-level observations rely on affinity reagents and lack appropriate genetic controls. Thus, evidence of a direct interaction remains elusive. We demonstrate that antibodies raised to endogenous human galectin-3 can isolate RNA-protein crosslinks, but this activity remains insensitive to LGALS3 knock-out. Proteomic characterization of anti-galectin-3 IPs revealed enrichment of galectin-3, but high abundance of hnRNPA2B1, an abundant, well-characterized RNA-binding protein with weak homology to the N-terminal domain of galectin-3, in the isolate. Genetic ablation of HNRNPA2B1, but not LGALS3, eliminates the ability of the anti-galectin-3 antibodies to isolate RNA-protein crosslinks, implying either an indirect interaction or cross-reactivity. To address this, we introduced an epitope tag to the endogenous C-terminal locus of LGALS3. Isolation of the tagged galectin-3 failed to reveal any RNA-protein crosslinks. This result suggests that the galectin-3 does not directly interact with RNA and may be misidentified as an RNA-binding protein, at least in HeLa where the putative RNA associations were first identified. We encourage further investigation of this phenomenon employ gene deletions and, when possible, endogenous epitope tags to achieve the specificity required to evaluate potential interactions.
Collapse
Affiliation(s)
- Egan L Peltan
- Department of Chemical and Systems Biology, Stanford University School of
Medicine, 269 Campus Drive CCSR 4145 Stanford, CA
94305, United States
- Sarafan ChEM-H, Stanford University, Stanford
ChEM-H Building 290 Jane Stanford Way Stanford, CA 94305, United States
| | - Nicholas M Riley
- Sarafan ChEM-H, Stanford University, Stanford
ChEM-H Building 290 Jane Stanford Way Stanford, CA 94305, United States
- Department of Chemistry, Stanford University, 333
Campus Drive Stanford, CA 94305, United
States
| | - Ryan A Flynn
- Stem Cell Program and Division of Hematology/Oncology,
Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA
02445, United States
- Department of Stem Cell and Regenerative Biology, Harvard
University, 7 Divinity Ave, Cambridge, MA 02138,
United States
| | - David S Roberts
- Sarafan ChEM-H, Stanford University, Stanford
ChEM-H Building 290 Jane Stanford Way Stanford, CA 94305, United States
- Department of Chemistry, Stanford University, 333
Campus Drive Stanford, CA 94305, United
States
| | - Carolyn R Bertozzi
- Sarafan ChEM-H, Stanford University, Stanford
ChEM-H Building 290 Jane Stanford Way Stanford, CA 94305, United States
- Department of Chemistry, Stanford University, 333
Campus Drive Stanford, CA 94305, United
States
- Howard Hughes Medical Institute, Stanford University,
279 Campus Drive Room B202 Stanford, CA 94305-5323, United States
| |
Collapse
|
35
|
Ping WX, Hu S, Su JQ, Ouyang SY. Metabolic disorders in prediabetes: From mechanisms to therapeutic management. World J Diabetes 2024; 15:361-377. [PMID: 38591088 PMCID: PMC10999048 DOI: 10.4239/wjd.v15.i3.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/04/2024] [Accepted: 02/07/2024] [Indexed: 03/15/2024] Open
Abstract
Diabetes, one of the world's top ten diseases, is known for its high mortality and complication rates and low cure rate. Prediabetes precedes the onset of diabetes, during which effective treatment can reduce diabetes risk. Prediabetes risk factors include high-calorie and high-fat diets, sedentary lifestyles, and stress. Consequences may include considerable damage to vital organs, including the retina, liver, and kidneys. Interventions for treating prediabetes include a healthy lifestyle diet and pharmacological treatments. However, while these options are effective in the short term, they may fail due to the difficulty of long-term implementation. Medications may also be used to treat prediabetes. This review examines prediabetic treatments, particularly metformin, glucagon-like peptide-1 receptor agonists, sodium glucose cotransporter 2 inhibitors, vitamin D, and herbal medicines. Given the remarkable impact of prediabetes on the progression of diabetes mellitus, it is crucial to intervene promptly and effectively to regulate prediabetes. However, the current body of research on prediabetes is limited, and there is considerable confusion surrounding clinically relevant medications. This paper aims to provide a comprehensive summary of the pathogenesis of pre-diabetes mellitus and its associated therapeutic drugs. The ultimate goal is to facilitate the clinical utilization of medications and achieve efficient and timely control of diabetes mellitus.
Collapse
Affiliation(s)
- Wen-Xin Ping
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, Fujian Province, China
| | - Shan Hu
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, Fujian Province, China
| | - Jing-Qian Su
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, Fujian Province, China
| | - Song-Ying Ouyang
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, Fujian Province, China
| |
Collapse
|
36
|
Behrooz AB, Cordani M, Donadelli M, Ghavami S. Metastatic outgrowth via the two-way interplay of autophagy and metabolism. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166824. [PMID: 37949196 DOI: 10.1016/j.bbadis.2023.166824] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 11/12/2023]
Abstract
Metastasis represents one of the most dangerous issue of cancer progression, characterized by intricate interactions between invading tumor cells, various proteins, and other cells on the way towards target sites. Tumor cells, while undergoing metastasis, engage in dynamic dialogues with stromal cells and undertake epithelial-mesenchymal transition (EMT) phenoconversion. To ensure survival, tumor cells employ several strategies such as restructuring their metabolic needs to adapt to the alterations of the microenvironmental resources via different mechanisms including macroautophagy (autophagy) and to circumvent anoikis-a form of cell death induced upon detachment from the extracellular matrix (ECM). This review focuses on the puzzling connections of autophagy and energetic metabolism within the context of cancer metastasis.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada; Academy of Silesia, Faculty of Medicine, Rolna 43 Street, 40-555 Katowice, Poland; Department of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
37
|
Chauhan N, Patro BS. Emerging roles of lysosome homeostasis (repair, lysophagy and biogenesis) in cancer progression and therapy. Cancer Lett 2024; 584:216599. [PMID: 38135207 DOI: 10.1016/j.canlet.2023.216599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
In the era of personalized therapy, precise targeting of subcellular organelles holds great promise for cancer modality. Taking into consideration that lysosome represents the intersection site in numerous endosomal trafficking pathways and their modulation in cancer growth, progression, and resistance against cancer therapies, the lysosome is proposed as an attractive therapeutic target for cancer treatment. Based on the recent advances, the current review provides a comprehensive understanding of molecular mechanisms of lysosome homeostasis under 3R responses: Repair, Removal (lysophagy) and Regeneration of lysosomes. These arms of 3R responses have distinct role in lysosome homeostasis although their interdependency along with switching between the pathways still remain elusive. Recent advances underpinning the crucial role of (1) ESCRT complex dependent/independent repair of lysosome, (2) various Galectins-based sensing and ubiquitination in lysophagy and (3) TFEB/TFE proteins in lysosome regeneration/biogenesis of lysosome are outlined. Later, we also emphasised how these recent advancements may aid in development of phytochemicals and pharmacological agents for targeting lysosomes for efficient cancer therapy. Some of these lysosome targeting agents, which are now at various stages of clinical trials and patents, are also highlighted in this review.
Collapse
Affiliation(s)
- Nitish Chauhan
- Bio-Organic Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, Maharashtra, 400094, India
| | - Birija Sankar Patro
- Bio-Organic Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, Maharashtra, 400094, India.
| |
Collapse
|
38
|
Shariq M, Khan MF, Raj R, Ahsan N, Kumar P. PRKAA2, MTOR, and TFEB in the regulation of lysosomal damage response and autophagy. J Mol Med (Berl) 2024; 102:287-311. [PMID: 38183492 DOI: 10.1007/s00109-023-02411-7] [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: 05/08/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/08/2024]
Abstract
Lysosomes function as critical signaling hubs that govern essential enzyme complexes. LGALS proteins (LGALS3, LGALS8, and LGALS9) are integral to the endomembrane damage response. If ESCRT fails to rectify damage, LGALS-mediated ubiquitination occurs, recruiting autophagy receptors (CALCOCO2, TRIM16, and SQSTM1) and VCP/p97 complex containing UBXN6, PLAA, and YOD1, initiating selective autophagy. Lysosome replenishment through biogenesis is regulated by TFEB. LGALS3 interacts with TFRC and TRIM16, aiding ESCRT-mediated repair and autophagy-mediated removal of damaged lysosomes. LGALS8 inhibits MTOR and activates TFEB for ATG and lysosomal gene transcription. LGALS9 inhibits USP9X, activates PRKAA2, MAP3K7, ubiquitination, and autophagy. Conjugation of ATG8 to single membranes (CASM) initiates damage repair mediated by ATP6V1A, ATG16L1, ATG12, ATG5, ATG3, and TECPR1. ATG8ylation or CASM activates the MERIT system (ESCRT-mediated repair, autophagy-mediated clearance, MCOLN1 activation, Ca2+ release, RRAG-GTPase regulation, MTOR modulation, TFEB activation, and activation of GTPase IRGM). Annexins ANAX1 and ANAX2 aid damage repair. Stress granules stabilize damaged membranes, recruiting FLCN-FNIP1/2, G3BP1, and NUFIP1 to inhibit MTOR and activate TFEB. Lysosomes coordinate the synergistic response to endomembrane damage and are vital for innate and adaptive immunity. Future research should unveil the collaborative actions of ATG proteins, LGALSs, TRIMs, autophagy receptors, and lysosomal proteins in lysosomal damage response.
Collapse
Affiliation(s)
- Mohd Shariq
- Quantlase Imaging Laboratory, Quantlase Lab LLC, Unit 1-8, Masdar City, Abu Dhabi, UAE.
| | - Mohammad Firoz Khan
- Quantlase Imaging Laboratory, Quantlase Lab LLC, Unit 1-8, Masdar City, Abu Dhabi, UAE.
| | - Reshmi Raj
- Quantlase Imaging Laboratory, Quantlase Lab LLC, Unit 1-8, Masdar City, Abu Dhabi, UAE
| | - Nuzhat Ahsan
- Quantlase Imaging Laboratory, Quantlase Lab LLC, Unit 1-8, Masdar City, Abu Dhabi, UAE
| | - Pramod Kumar
- Quantlase Imaging Laboratory, Quantlase Lab LLC, Unit 1-8, Masdar City, Abu Dhabi, UAE
| |
Collapse
|
39
|
Troncoso MF, Elola MT, Blidner AG, Sarrias L, Espelt MV, Rabinovich GA. The universe of galectin-binding partners and their functions in health and disease. J Biol Chem 2023; 299:105400. [PMID: 37898403 PMCID: PMC10696404 DOI: 10.1016/j.jbc.2023.105400] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/30/2023] Open
Abstract
Galectins, a family of evolutionarily conserved glycan-binding proteins, play key roles in diverse biological processes including tissue repair, adipogenesis, immune cell homeostasis, angiogenesis, and pathogen recognition. Dysregulation of galectins and their ligands has been observed in a wide range of pathologic conditions including cancer, autoimmune inflammation, infection, fibrosis, and metabolic disorders. Through protein-glycan or protein-protein interactions, these endogenous lectins can shape the initiation, perpetuation, and resolution of these processes, suggesting their potential roles in disease monitoring and treatment. However, despite considerable progress, a full understanding of the biology and therapeutic potential of galectins has not been reached due to their diversity, multiplicity of cell targets, and receptor promiscuity. In this article, we discuss the multiple galectin-binding partners present in different cell types, focusing on their contributions to selected physiologic and pathologic settings. Understanding the molecular bases of galectin-ligand interactions, particularly their glycan-dependency, the biochemical nature of selected receptors, and underlying signaling events, might contribute to designing rational therapeutic strategies to control a broad range of pathologic conditions.
Collapse
Affiliation(s)
- María F Troncoso
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María T Elola
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ada G Blidner
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Luciana Sarrias
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María V Espelt
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
40
|
Wang K, Fu S, Dong L, Zhang D, Wang M, Wu X, Shen E, Luo L, Li C, Nice EC, Huang C, Zou B. Periplocin suppresses the growth of colorectal cancer cells by triggering LGALS3 (galectin 3)-mediated lysophagy. Autophagy 2023; 19:3132-3150. [PMID: 37471054 PMCID: PMC10621285 DOI: 10.1080/15548627.2023.2239042] [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: 07/05/2022] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide and remains a major clinical challenge. Periplocin, a major bioactive component of the traditional Chinese herb Cortex periplocae, has recently been reported to be a potential anticancer drug. However, the mechanism of action is poorly understood. Here, we show that periplocin exhibits promising anticancer activity against CRC both in vitro and in vivo. Mechanistically, periplocin promotes lysosomal damage and induces apoptosis in CRC cells. Notably, periplocin upregulates LGALS3 (galectin 3) by binding and preventing LGALS3 from Lys210 ubiquitination-mediated proteasomal degradation, leading to the induction of excessive lysophagy and resultant exacerbation of lysosomal damage. Inhibition of LGALS3-mediated lysophagy attenuates periplocin-induced lysosomal damage and growth inhibition in CRC cells, suggesting a critical role of lysophagy in the anticancer effects of periplocin. Taken together, our results reveal a novel link between periplocin and the lysophagy machinery, and indicate periplocin as a potential therapeutic option for the treatment of CRC.Abbreviations: 3-MA: 3-methyladenine; ACACA/ACC1: acetyl-CoA carboxylase alpha; AMPK: adenosine monophosphate-activated protein kinase; AO: Acridine orange; ATG5: autophagy related 5; ATG7: autophagy related 7; CALM: calmodulin; CHX: cycloheximide; CRC: colorectal cancer; CQ: chloroquine; CTSB: cathepsin B; CTSD: cathepsin D; ESCRT: endosomal sorting complex required for transport; LAMP1: lysosomal associated membrane protein 1; LMP: lysosomal membrane permeabilization; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MCOLN1/TRPML1: mucolipin TRP cation channel 1; MKI67/Ki-67: marker of proliferation Ki-67; MTOR: mechanistic target of rapamycin kinase; P2RX4/P2X4: purinergic receptor P2X 4; PARP1/PARP: poly(ADP-ribose) polymerase 1; PRKAA/AMPKα: protein kinase AMP-activated catalytic subunit alpha; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; TRIM16: tripartite motif containing 16.
Collapse
Affiliation(s)
- Kui Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Shuyue Fu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Lixia Dong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Dingyue Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Mao Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xingyun Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Enhao Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Li Luo
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, West China Second University Hospital, Chengdu, Sichuan, P. R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, Sichuan, P. R. China
| | - Changlong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Edouard Collins Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Bingwen Zou
- Department of Thoracic Oncology and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
41
|
Boya P, Kaarniranta K, Handa JT, Sinha D. Lysosomes in retinal health and disease. Trends Neurosci 2023; 46:1067-1082. [PMID: 37848361 PMCID: PMC10842632 DOI: 10.1016/j.tins.2023.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/06/2023] [Accepted: 09/24/2023] [Indexed: 10/19/2023]
Abstract
Lysosomes play crucial roles in various cellular processes - including endocytosis, phagocytosis, and autophagy - which are vital for maintaining retinal health. Moreover, these organelles serve as environmental sensors and act as central hubs for multiple signaling pathways. Through communication with other cellular components, such as mitochondria, lysosomes orchestrate the cytoprotective response essential for preserving cellular homeostasis. This coordination is particularly critical in the retina, given its high metabolic rate and susceptibility to photo-oxidative stress. Consequently, impaired lysosomal function and dysregulated communication between lysosomes and other organelles contribute significantly to the pathobiology of major retinal degenerative diseases. This review explores the pivotal role of lysosomes in retinal cells and their involvement in retinal degenerative diseases.
Collapse
Affiliation(s)
- Patricia Boya
- Department of Neuroscience, University of Fribourg, Fribourg, Switzerland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland; Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - James T Handa
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Debasish Sinha
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
42
|
Chae CW, Yoon JH, Lim JR, Park JY, Cho JH, Jung YH, Choi GE, Lee HJ, Han HJ. TRIM16-mediated lysophagy suppresses high-glucose-accumulated neuronal Aβ. Autophagy 2023; 19:2752-2768. [PMID: 37357416 PMCID: PMC10472864 DOI: 10.1080/15548627.2023.2229659] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023] Open
Abstract
ABBREVIATIONS Aβ: amyloid β; AD: Alzheimer disease; AMPK: 5' adenosine monophosphate-activated protein kinase; CTSB: cathepsin B; CTSD: cathepsin D; DM: diabetes mellitus; ESCRT: endosomal sorting complex required for transport; FBXO27: F-box protein 27; iPSC-NDs: induced pluripotent stem cell-derived neuronal differentiated cells; LAMP1: lysosomal-associated membrane protein 1; LMP: lysosomal membrane permeabilization; LRSAM1: leucine rich repeat and sterile alpha motif containing 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; p-MAPT/tau: phosphorylated microtubule associated protein tau; ROS: reactive oxygen species; STZ: streptozotocin; TFE3: transcription factor E3; TFEB: transcription factor EB; TRIM16: tripartite motif containing 16; UBE2QL1: ubiquitin conjugating enzyme E2 Q family like 1; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Jee Hyeon Yoon
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Jae Ryong Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ji Yong Park
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ji Hyeon Cho
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Gee Euhn Choi
- Laboratory of Veterinary Biochemistry, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, South Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, South Korea
| | - Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, South Korea
- Institute for Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| |
Collapse
|
43
|
Gong J, Liu Y, Wang W, He R, Xia Q, Chen L, Zhao C, Gao Y, Shi Y, Bai Y, Liao Y, Zhang Q, Zhu F, Wang M, Li X, Qin R. TRIM21-Promoted FSP1 Plasma Membrane Translocation Confers Ferroptosis Resistance in Human Cancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302318. [PMID: 37587773 PMCID: PMC10582465 DOI: 10.1002/advs.202302318] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/21/2023] [Indexed: 08/18/2023]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by excessive accumulation of lipid peroxides, has become a promising strategy in cancer treatment. Cancer cells exploit antioxidant proteins, including Ferroptosis Suppressor Protein 1 (FSP1), to prevent ferroptosis. In this study, it is found that the E3 ubiquitin ligase TRIM21 bound to FSP1 and mediated its ubiquitination on K322 and K366 residues via K63 linkage, which is essential for its membrane translocation and ferroptosis suppression ability. It is further verified the protective role of the TRIM21-FSP1 axis in RSL3-induced ferroptosis in cancer cells and a subcutaneous tumor model. Moreover, TRIM21 is highly expressed in multiple gastrointestinal (GI) tumors, and its expression is further stimulated upon ferroptosis induction in cancer cells and the KPC mouse model. In summary, This study identifies TRIM21 as a negative regulator of ferroptosis through K63 ubiquitination of FSP1, which can serve as a therapeutic target to enhance the chemosensitivity of tumors based on ferroptosis induction.
Collapse
Affiliation(s)
- Jun Gong
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Yuhui Liu
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Wenjia Wang
- Institute of Integrated Traditional Chinese and Western MedicineAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Ruizhi He
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Qilong Xia
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Lin Chen
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Chunle Zhao
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Yang Gao
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Yongkang Shi
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Yu Bai
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Yangwei Liao
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Qi Zhang
- Department of Plastic and Cosmetic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Feng Zhu
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Min Wang
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Xu Li
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| | - Renyi Qin
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhanHubei430030China
| |
Collapse
|
44
|
Nicolao MC, Rodrigues CR, Coccimiglio MB, Ledo C, Docena GH, Cumino AC. Characterization of protein cargo of Echinococcus granulosus extracellular vesicles in drug response and its influence on immune response. Parasit Vectors 2023; 16:255. [PMID: 37516852 PMCID: PMC10387209 DOI: 10.1186/s13071-023-05854-6] [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: 03/14/2023] [Accepted: 06/28/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND The Echinococcus granulosus sensu lato species complex causes cystic echinococcosis, a zoonotic disease of medical importance. Parasite-derived small extracellular vesicles (sEVs) are involved in the interaction with hosts intervening in signal transduction related to parasite proliferation and disease pathogenesis. Although the characteristics of sEVs from E. granulosus protoscoleces and their interaction with host dendritic cells (DCs) have been described, the effect of sEVs recovered during parasite pharmacological treatment on the immune response remains unexplored. METHODS Here, we isolated and characterized sEVs from control and drug-treated protoscoleces by ultracentrifugation, transmission electron microscopy, dynamic light scattering, and proteomic analysis. In addition, we evaluated the cytokine response profile induced in murine bone marrow-derived dendritic cells (BMDCs) by qPCR. RESULTS The isolated sEVs, with conventional size between 50 and 200 nm, regardless of drug treatment, showed more than 500 cargo proteins and, importantly, 20 known antigens and 70 potential antigenic proteins, and several integral-transmembrane and soluble proteins mainly associated with signal transduction, immunomodulation, scaffolding factors, extracellular matrix-anchoring, and lipid transport. The identity and abundance of proteins in the sEV-cargo from metformin- and albendazole sulfoxide (ABZSO)-treated parasites were determined by proteomic analysis, detecting 107 and eight exclusive proteins, respectively, which include proteins related to the mechanisms of drug action. We also determined that the interaction of murine BMDCs with sEVs derived from control parasites and those treated with ABZSO and metformin increased the expression of pro-inflammatory cytokines such as IL-12 compared to control cells. Additionally, protoscolex-derived vesicles from metformin treatments induced the production of IL-6, TNF-α, and IL-10. However, the expression of IL-23 and TGF-β was downregulated. CONCLUSIONS We demonstrated that sEV-cargo derived from drug-treated E. granulosus protoscoleces have immunomodulatory functions, as they enhance DC activation towards a type 1 pro-inflammatory profile against the parasite, and therefore support the proposal of a new approach for the prevention and treatment of secondary echinococcosis.
Collapse
Affiliation(s)
- María Celeste Nicolao
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Christian Rodriguez Rodrigues
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, 7600, Mar del Plata, Argentina
| | - Magalí B Coccimiglio
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina
| | - Camila Ledo
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina
| | - Guillermo H Docena
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), La Plata, Argentina
| | - Andrea C Cumino
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, 7600, Mar del Plata, Argentina.
| |
Collapse
|
45
|
Qian H, Lu Z, Hao C, Zhao Y, Bo X, Hu Y, Zhang Y, Yao Y, Ma G, Chen L. TRIM44 aggravates cardiac fibrosis after myocardial infarction via TAK1 stabilization. Cell Signal 2023:110744. [PMID: 37271349 DOI: 10.1016/j.cellsig.2023.110744] [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: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/29/2023] [Indexed: 06/06/2023]
Abstract
Myocardial infarction (MI) is one of the most dangerous cardiovascular events. Cardiac fibrosis is a common pathological feature of remodeling after injury that is related to adverse clinical results with no effective treatment. Previous studies have confirmed that TRIM44, an E3 ligase, can promote the proliferation and migration of various tumor cells. However, the role of TRIM44 in cardiac fibrosis remains unknown. Models of TGF-β1 stimulation and MI-induced fibrosis were established to investigate the role and potential underlying mechanism of TRIM44 in cardiac fibrosis. The results showed that cardiac fibrosis was significantly inhibited after TRIM44 knockdown in a mouse model of MI, while it was enhanced when TRIM44 was overexpressed. Furthermore, in vitro studies showed that fibrosis markers were significantly reduced in cardiac fibroblasts (CFs) with TRIM44 knockdown, whereas TRIM44 overexpression promoted the expression of fibrosis markers. Mechanistically, TRIM44 maintains TAK1 stability by inhibiting the degradation of k48-linked polyubiquitination-mediated ubiquitination, thereby increasing phosphorylated TAK1 expression in the fibrotic environment and activating MAPKs to promote fibrosis. Pharmacological inhibition of TAK1 phosphorylation reversed the fibrogenic effects of TRIM44 overexpression. Combined, these results suggest that TRIM44 is a potential therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Hao Qian
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhengri Lu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chunshu Hao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiangwei Bo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ya Hu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Nanjing 211200, China.
| |
Collapse
|
46
|
Morrison HM, Craft J, Rivera-Lugo R, Johnson JR, Golovkine GR, Bell SL, Dodd CE, Van Dis E, Beatty WL, Margolis SR, Repasy T, Shaker I, Lee AY, Vance RE, Stanley SA, Watson RO, Krogan NJ, Portnoy DA, Penn BH, Cox JS. Deficiency in Galectin-3, -8, and -9 impairs immunity to chronic Mycobacterium tuberculosis infection but not acute infection with multiple intracellular pathogens. PLoS Pathog 2023; 19:e1011088. [PMID: 37352334 PMCID: PMC10325092 DOI: 10.1371/journal.ppat.1011088] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 07/06/2023] [Accepted: 05/01/2023] [Indexed: 06/25/2023] Open
Abstract
Macrophages employ an array of pattern recognition receptors to detect and eliminate intracellular pathogens that access the cytosol. The cytosolic carbohydrate sensors Galectin-3, -8, and -9 (Gal-3, Gal-8, and Gal-9) recognize damaged pathogen-containing phagosomes, and Gal-3 and Gal-8 are reported to restrict bacterial growth via autophagy in cultured cells. However, the contribution of these galectins to host resistance during bacterial infection in vivo remains unclear. We found that Gal-9 binds directly to Mycobacterium tuberculosis (Mtb) and Salmonella enterica serovar Typhimurium (Stm) and localizes to Mtb in macrophages. To determine the combined contribution of membrane damage-sensing galectins to immunity, we generated Gal-3, -8, and -9 triple knockout (TKO) mice. Mtb infection of primary macrophages from TKO mice resulted in defective autophagic flux but normal bacterial replication. Surprisingly, these mice had no discernable defect in resistance to acute infection with Mtb, Stm or Listeria monocytogenes, and had only modest impairments in bacterial growth restriction and CD4 T cell activation during chronic Mtb infection. Collectively, these findings indicate that while Gal-3, -8, and -9 respond to an array of intracellular pathogens, together these membrane damage-sensing galectins play a limited role in host resistance to bacterial infection.
Collapse
Affiliation(s)
- Huntly M. Morrison
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Julia Craft
- Department of Internal Medicine, Division of Infectious Diseases, University of California, Davis, Davis, California, United States of America
| | - Rafael Rivera-Lugo
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Jeffery R. Johnson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco; Quantitative Biosciences Institute (QBI), University of California, San Francisco; Gladstone Institutes, San Francisco, California, United States of America
| | - Guillaume R. Golovkine
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Samantha L. Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, School of Medicine, Bryan, Texas, United States of America
| | - Claire E. Dodd
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Erik Van Dis
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Shally R. Margolis
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Teresa Repasy
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Isaac Shaker
- Department of Internal Medicine, Division of Infectious Diseases, University of California, Davis, Davis, California, United States of America
| | - Angus Y. Lee
- Cancer Research Laboratory, University of California, Berkeley, Berkeley, California, United States of America
| | - Russell E. Vance
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Sarah A. Stanley
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
- School of Public Health, Division of Infectious Diseases and Vaccinology, University of California, Berkeley, Berkeley, California, United States of America
| | - Robert O. Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, School of Medicine, Bryan, Texas, United States of America
| | - Nevan J. Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco; Quantitative Biosciences Institute (QBI), University of California, San Francisco; Gladstone Institutes, San Francisco, California, United States of America
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Bennett H. Penn
- Department of Internal Medicine, Division of Infectious Diseases, University of California, Davis, Davis, California, United States of America
| | - Jeffery S. Cox
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| |
Collapse
|
47
|
Wang F, Peters R, Jia J, Mudd M, Salemi M, Allers L, Javed R, Duque TLA, Paddar MA, Trosdal ES, Phinney B, Deretic V. ATG5 provides host protection acting as a switch in the atg8ylation cascade between autophagy and secretion. Dev Cell 2023; 58:866-884.e8. [PMID: 37054706 PMCID: PMC10205698 DOI: 10.1016/j.devcel.2023.03.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/26/2023] [Accepted: 03/20/2023] [Indexed: 04/15/2023]
Abstract
ATG5 is a part of the E3 ligase directing lipidation of ATG8 proteins, a process central to membrane atg8ylation and canonical autophagy. Loss of Atg5 in myeloid cells causes early mortality in murine models of tuberculosis. This in vivo phenotype is specific to ATG5. Here, we show using human cell lines that absence of ATG5, but not of other ATGs directing canonical autophagy, promotes lysosomal exocytosis and secretion of extracellular vesicles and, in murine Atg5fl/fl LysM-Cre neutrophils, their excessive degranulation. This is due to lysosomal disrepair in ATG5 knockout cells and the sequestration by an alternative conjugation complex, ATG12-ATG3, of ESCRT protein ALIX, which acts in membrane repair and exosome secretion. These findings reveal a previously undescribed function of ATG5 in its host-protective role in murine experimental models of tuberculosis and emphasize the significance of the branching aspects of the atg8ylation conjugation cascade beyond the canonical autophagy.
Collapse
Affiliation(s)
- Fulong Wang
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ryan Peters
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Michal Mudd
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Michelle Salemi
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ruheena Javed
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Thabata L A Duque
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Masroor A Paddar
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Einar S Trosdal
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Brett Phinney
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA.
| |
Collapse
|
48
|
Golovkine GR, Roberts AW, Morrison HM, Rivera-Lugo R, McCall RM, Nilsson H, Garelis NE, Repasy T, Cronce M, Budzik J, Van Dis E, Popov LM, Mitchell G, Zalpuri R, Jorgens D, Cox JS. Autophagy restricts Mycobacterium tuberculosis during acute infection in mice. Nat Microbiol 2023; 8:819-832. [PMID: 37037941 PMCID: PMC11027733 DOI: 10.1038/s41564-023-01354-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/03/2023] [Indexed: 04/12/2023]
Abstract
Whether or not autophagy has a role in defence against Mycobacterium tuberculosis infection remains unresolved. Previously, conditional knockdown of the core autophagy component ATG5 in myeloid cells was reported to confer extreme susceptibility to M. tuberculosis in mice, whereas depletion of other autophagy factors had no effect on infection. We show that doubling cre gene dosage to more robustly deplete ATG16L1 or ATG7 resulted in increased M. tuberculosis growth and host susceptibility in mice, although ATG5-depleted mice are more sensitive than ATG16L1- or ATG7-depleted mice. We imaged individual macrophages infected with M. tuberculosis and identified a shift from apoptosis to rapid necrosis in autophagy-depleted cells. This effect was dependent on phagosome permeabilization by M. tuberculosis. We monitored infected cells by electron microscopy, showing that autophagy protects the host macrophage by partially reducing mycobacterial access to the cytosol. We conclude that autophagy has an important role in defence against M. tuberculosis in mammals.
Collapse
Affiliation(s)
- Guillaume R Golovkine
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Evotec, Toulouse, France
| | - Allison W Roberts
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Huntly M Morrison
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Rafael Rivera-Lugo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Rita M McCall
- Department of Plant & Microbial Biology, University of California, Berkeley, CA, USA
| | - Hannah Nilsson
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Nicholas E Garelis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Teresa Repasy
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Bio-Rad Laboratories, Seattle, WA, USA
| | - Michael Cronce
- Department of Bioengineering, University of California, Berkeley, CA, USA
- UC Berkeley-UCSF Graduate program in Bioengineering, Berkeley, CA, USA
| | - Jonathan Budzik
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Erik Van Dis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Lauren M Popov
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Novome Biotechnologies, San Francisco, CA, USA
| | - Gabriel Mitchell
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Open Innovation @ NITD, Novartis Institute for Tropical Diseases, Emeryville, CA, USA
| | - Reena Zalpuri
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Danielle Jorgens
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Jeffery S Cox
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
49
|
Pełech A, Ruszała M, Niebrzydowska-Tatus M, Bień K, Kimber-Trojnar Ż, Czuba M, Świstowska M, Leszczyńska-Gorzelak B. Do Serum Galectin-9 Levels in Women with Gestational Diabetes and Healthy Ones Differ before or after Delivery? A Pilot Study. Biomolecules 2023; 13:biom13040697. [PMID: 37189444 DOI: 10.3390/biom13040697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is a common metabolic disease that occurs during pregnancy, with the placenta playing an important role in its pathophysiology. Currently, the role of galectin-9 in the development of GDM is unknown. The aim of this study was to compare galectin-9 concentrations in healthy pregnant women and those with GDM. Galectin-9 levels were assessed in serum samples taken both just before and after delivery, as well as in urine samples collected in the postpartum period. Maternal body composition and hydration status were evaluated using the bioelectrical impedance analysis (BIA) method. There were no statistically significant differences in the concentration of galectin-9 in women with GDM compared to healthy pregnant women in their serum samples taken just before delivery, nor in their serum and urine samples collected in the early postpartum period. However, serum galectin-9 concentrations taken before delivery were positively correlated with BMI and parameters related to the amount of adipose tissue assessed in the early postpartum period. Additionally, there was a correlation between serum galectin-9 concentrations taken before and after delivery. Galectin-9 is unlikely to become a diagnostic marker for GDM. However, this subject requires further clinical research in larger groups.
Collapse
Affiliation(s)
- Aleksandra Pełech
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Monika Ruszała
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | | | - Katarzyna Bień
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Żaneta Kimber-Trojnar
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Monika Czuba
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland
| | - Małgorzata Świstowska
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland
| | | |
Collapse
|
50
|
Steinberg GR, Hardie DG. New insights into activation and function of the AMPK. Nat Rev Mol Cell Biol 2023; 24:255-272. [PMID: 36316383 DOI: 10.1038/s41580-022-00547-x] [Citation(s) in RCA: 396] [Impact Index Per Article: 198.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
The classical role of AMP-activated protein kinase (AMPK) is as a cellular energy sensor activated by falling energy status, signalled by increases in AMP to ATP and ADP to ATP ratios. Once activated, AMPK acts to restore energy homeostasis by promoting ATP-producing catabolic pathways while inhibiting energy-consuming processes. In this Review, we provide an update on this canonical (AMP/ADP-dependent) activation mechanism, but focus mainly on recently described non-canonical pathways, including those by which AMPK senses the availability of glucose, glycogen or fatty acids and by which it senses damage to lysosomes and nuclear DNA. We also discuss new findings on the regulation of carbohydrate and lipid metabolism, mitochondrial and lysosomal homeostasis, and DNA repair. Finally, we discuss the role of AMPK in cancer, obesity, diabetes, nonalcoholic steatohepatitis (NASH) and other disorders where therapeutic targeting may exert beneficial effects.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|