1
|
Münz C, Campbell GR, Esclatine A, Faure M, Labonte P, Lussignol M, Orvedahl A, Altan-Bonnet N, Bartenschlager R, Beale R, Cirone M, Espert L, Jung J, Leib D, Reggiori F, Sanyal S, Spector SA, Thiel V, Viret C, Wei Y, Wileman T, Wodrich H. Autophagy machinery as exploited by viruses. AUTOPHAGY REPORTS 2025; 4:27694127.2025.2464986. [PMID: 40201908 PMCID: PMC11921968 DOI: 10.1080/27694127.2025.2464986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
Viruses adapt and modulate cellular pathways to allow their replication in host cells. The catabolic pathway of macroautophagy, for simplicity referred to as autophagy, is no exception. In this review, we discuss anti-viral functions of both autophagy and select components of the autophagy machinery, and how viruses have evaded them. Some viruses use the membrane remodeling ability of the autophagy machinery to build their replication compartments in the cytosol or efficiently egress from cells in a non-lytic fashion. Some of the autophagy machinery components and their remodeled membranes can even be found in viral particles as envelopes or single membranes around virus packages that protect them during spreading and transmission. Therefore, studies on autophagy regulation by viral infections can reveal functions of the autophagy machinery beyond lysosomal degradation of cytosolic constituents. Furthermore, they can also pinpoint molecular interactions with which the autophagy machinery can most efficiently be manipulated, and this may be relevant to develop effective disease treatments based on autophagy modulation.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich Switzerland
| | - Grant R Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of SD, Vermillion, SD, USA
| | - Audrey Esclatine
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Patrick Labonte
- eINRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada
| | - Marion Lussignol
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anthony Orvedahl
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Bartenschlager
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division Virus-Associated Carcinogenesis, Heidelberg, Germany
- German Centre for Infection Research, Heidelberg partner site, Heidelberg, Germany
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
- Division of Medicine, University College London, London, UK
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucile Espert
- University of Montpellier, Montpellier, France
- CNRS, Institut de Recherche enInfectiologie deMontpellier (IRIM), Montpellier, France
| | - Jae Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - David Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH, USA
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, Aarhus C, Denmark
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, UK
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital, San Diego, CA, USA
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland, and Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Yu Wei
- Institut Pasteur-Theravectys Joint Laboratory, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia
- Quadram Institute Bioscience, Norwich Research Park, Norfolk, UK
| | - Harald Wodrich
- sLaboratoire de Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
2
|
Kimura Y, Kimura M, Miura N, Yoshino Y, Kono H. NOD1 deficiency promotes inflammation via autophagic degradation of ASK1. Commun Biol 2025; 8:781. [PMID: 40399666 PMCID: PMC12095521 DOI: 10.1038/s42003-025-08213-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) is a pattern recognition receptor of bacterial peptidoglycans. NOD1 facilitates the elimination of invading intracellular bacteria via autophagy induction. Here, we demonstrate that NOD1 exerts an anti-inflammatory effect mediated via the selective autophagy of host cell protein. In our study of Candida albicans water-soluble fraction (CAWS)-induced coronary arteritis, which is a mouse model of Kawasaki disease, we observed an exacerbated disease phenotype in NOD1-deficient mice. NOD1 deficiency induced a higher expression of inflammatory cytokines via CAWS and CAWS-induced endoplasmic reticulum (ER) stress in bone marrow-derived dendritic cells. Furthermore, exaggerated inflammation was dependent on apoptosis signal-regulated kinase 1 (ASK1). Notably, NOD1 directly interacted with ASK1, inducing selective autophagy of ASK1, which was dependent on ATG16L1, and thus competitively inhibiting ER stress-dependent ASK1 activation. Altogether, these results show that NOD1 modulates excessive inflammatory responses through the upregulation of autophagy.
Collapse
Affiliation(s)
- Yoshitaka Kimura
- Department of Microbiology and Immunology, Teikyo University School of Medicine, Tokyo, Japan.
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan.
| | - Miyako Kimura
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
- Division of Regenerative Therapy, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Noriko Miura
- Center for the Advancement of Pharmaceutical Education, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Yusuke Yoshino
- Department of Microbiology and Immunology, Teikyo University School of Medicine, Tokyo, Japan
- Asia International Institute of Infectious Disease Control, Teikyo University, Tokyo, Japan
| | - Hajime Kono
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
3
|
Yao X, Rudensky E, Martin PK, Miller BM, Vargas I, Zwack EE, Lacey KA, He Z, Furtado GC, Lira SA, Torres VJ, Shopsin B, Cadwell K. Heterozygosity for Crohn's disease risk allele of ATG16L1 promotes unique protein interactions and protects against bacterial infection. Immunity 2025:S1074-7613(25)00186-4. [PMID: 40373771 DOI: 10.1016/j.immuni.2025.04.023] [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: 03/08/2024] [Revised: 11/25/2024] [Accepted: 04/17/2025] [Indexed: 05/17/2025]
Abstract
The T300A substitution in ATG16L1 associated with Crohn's disease impairs autophagy, yet up to 50% of humans are heterozygous for this allele. Here, we demonstrate that heterozygosity for the analogous substitution in mice (Atg16L1T316A), but not homozygosity, protects against lethal Salmonella enterica Typhimurium infection. One copy of Atg16L1T316A was sufficient to enhance cytokine production through inflammasome activation, which was necessary for protection. In contrast, two copies of Atg16L1T316A inhibited the autophagy-related process of LC3-associated phagocytosis (LAP) and increased susceptibility. Macrophages from human donors heterozygous for ATG16L1T300A displayed elevated inflammasome activation while homozygosity impaired LAP, similar to mice. These results clarify how the T300A substitution impacts ATG16L1 function and suggest it can be beneficial to heterozygous carriers, providing an explanation for its prevalence within the human population.
Collapse
Affiliation(s)
- Xiaomin Yao
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Eugene Rudensky
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Patricia K Martin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Brittany M Miller
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Isabel Vargas
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Erin E Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Zhengxiang He
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Glaucia C Furtado
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sérgio A Lira
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, New York, NY 10016, USA; Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, New York, NY 10016, USA; Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
4
|
Zhang M, Wu C, Lu D, Wang X, Shang G. cGAS-STING: mechanisms and therapeutic opportunities. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1309-1323. [PMID: 39821837 DOI: 10.1007/s11427-024-2808-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025]
Abstract
The cGAS-STING pathway plays a crucial role in the innate immune system by detecting mislocalized double-stranded DNA (dsDNA) in the cytoplasm and triggering downstream signal transduction. Understanding the mechanisms by which cGAS and STING operate is vital for gaining insights into the biology of this pathway. This review provides a detailed examination of the structural features of cGAS and STING proteins, with a particular emphasis on their activation and inhibition mechanisms. We also discuss the novel discovery of STING functioning as an ion channel. Furthermore, we offer an overview of key agonists and antagonists of cGAS and STING, shedding light on their mechanisms of action. Deciphering the molecular intricacies of the cGAS-STING pathway holds significant promise for the development of targeted therapies aimed at maintaining immune homeostasis within both innate and adaptive immunity.
Collapse
Affiliation(s)
- Mengyuan Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Changxin Wu
- The Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Defen Lu
- College of Life Sciences, Shanxi Agricultural University, Taiyuan, 030031, China.
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China.
| | - Guijun Shang
- The Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
- College of Life Sciences, Shanxi Agricultural University, Taiyuan, 030031, China.
| |
Collapse
|
5
|
Bone B, Griffith L, Jefferson M, Yamauchi Y, Wileman T, Powell PP. ATG16L1 WD domain and linker regulates lipid trafficking to maintain plasma membrane integrity to limit influenza virus infection. Autophagy 2025:1-16. [PMID: 40143422 DOI: 10.1080/15548627.2025.2482516] [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: 08/28/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
The non-canonical functions of autophagy protein ATG16L1 are dependent on a C-terminal WD domain. Recent studies show that the WD domain is required for conjugation of LC3 to single membranes during endocytosis and phagocytosis, where it is thought to promote fusion with lysosomes. Studies in cells lacking the WD domain suggest additional roles in the regulation of cytokine receptor recycling and plasma membrane repair. The WD domain also protects mice against lethal influenza virus in vivo. Here, analysis of mice lacking the WD domain (ΔWD) shows enrichment of cholesterol in brain tissue suggesting a role for the WD domain in cholesterol transport. Brain tissue and cells from ΔWD mice showed reduced cholesterol and phosphatidylserine (PS) in the plasma membrane. Cells from ΔWD mice also showed an intracellular accumulation of cholesterol predominantly in late endosomes. Infection studies using IAV suggest that the loss of cholesterol and PS from the plasma membrane in cells from ΔWD mice results in increased endocytosis and nuclear delivery of IAV, as well as increased Ifnb/Ifnβ and Isg15 gene expression. Upregulation of Il6, Ifnb and Isg15 mRNA were observed in "ex vivo" precision cut lung slices from ΔWD mice both at rest and in response to IAV infection. Overall, we present evidence that regulation of lipid transport by the WD domain of ATG16L1 may have downstream implications in attenuating viral infection and limiting lethal cytokine signaling.Abbreviations: BMDM: bone marrow-derived macrophages, CASM: conjugation of ATG8 to single membranes, CCD: coil-coil domain, IAV: influenza virus A, IFIT1: interferon-induced protein with tetratricopeptide repeats 1, IFITM3: interferon induced transmembrane protein 3, IFN: interferon, ISG15: ISG15 ubiquitin-like modifier, LANDO: LC3-associated endocytosis, LAP: LC3-associated phagocytosis, LDL: low density lipoprotein, NP: nucleoprotein, PS: phosphatidylserine, WD: WD40-repeat-containing C-terminal domain, WT: wild type.
Collapse
Affiliation(s)
- Benjamin Bone
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich, Norfolk, UK
| | - Luke Griffith
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich, Norfolk, UK
| | - Matthew Jefferson
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich, Norfolk, UK
| | - Yohei Yamauchi
- Molecular Medicine Laboratory, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zurich, Zurich, Switzerland
- Department of Virology, Graduate School of Medicine, Nagoya University, Japan
| | - Thomas Wileman
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich, Norfolk, UK
| | - Penny P Powell
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich, Norfolk, UK
| |
Collapse
|
6
|
Figueras-Novoa C, Akutsu M, Murata D, Weston A, Jiang M, Montaner B, Dubois C, Shenoy A, Beale R. Caspase cleavage of influenza A virus M2 disrupts M2-LC3 interaction and regulates virion production. EMBO Rep 2025; 26:1768-1791. [PMID: 40033051 PMCID: PMC11977235 DOI: 10.1038/s44319-025-00388-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Influenza A virus (IAV) Matrix 2 protein (M2) is an ion channel, required for efficient viral entry and egress. M2 interacts with the small ubiquitin-like LC3 protein through a cytoplasmic C-terminal LC3-interacting region (LIR). Here, we report that M2 is cleaved by caspases, abolishing the M2-LC3 interaction. A crystal structure of the M2 LIR in complex with LC3 indicates the caspase cleavage tetrapeptide motif (82SAVD85) is an unstructured linear motif that does not overlap with the LIR. IAV mutant expressing a permanently truncated M2, mimicking caspase cleavage, exhibit defects in M2 plasma membrane transport, viral filament formation, and virion production. Our results reveal a dynamic regulation of the M2-LC3 interaction by caspases. This highlights the role of host proteases in regulating IAV exit, relating virion production with host cell state.
Collapse
Affiliation(s)
- Carmen Figueras-Novoa
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
- Faculty of Life Sciences, University College London, London, UK
| | - Masato Akutsu
- Buchmann Institute for Molecular Life Sciences, Institute of Biochemistry II, Goethe University, Max-von Laue-Str. 15, Frankfurt, 60438, Germany
- Research Center for Advanced Analysis, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Daichi Murata
- Buchmann Institute for Molecular Life Sciences, Institute of Biochemistry II, Goethe University, Max-von Laue-Str. 15, Frankfurt, 60438, Germany
- Department of Biomolecular Chemistry, Kyoto Prefectural University, Hangi-cho, Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
- China Innovation Center, Shiseido China Co., Ltd., Shanghai, China
| | - Anne Weston
- Electron Microscopy STP, The Francis Crick Institute, London, UK
| | - Ming Jiang
- High Throughput Screening STP, The Francis Crick Institute, London, UK
| | - Beatriz Montaner
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
| | | | - Avinash Shenoy
- Department of Infectious Disease, Imperial College, London, UK.
- Satellite Group Leader, The Francis Crick Institute, London, UK.
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK.
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
7
|
Chen D, Fearns A, Gutierrez MG. Mycobacterium tuberculosis phagosome Ca 2+ leakage triggers multimembrane ATG8/LC3 lipidation to restrict damage in human macrophages. SCIENCE ADVANCES 2025; 11:eadt3311. [PMID: 40138395 PMCID: PMC11939036 DOI: 10.1126/sciadv.adt3311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025]
Abstract
The role of canonical autophagy in controlling Mycobacterium tuberculosis (Mtb), referred to as xenophagy, is understood to involve targeting Mtb to autophagosomes, which subsequently fuse with lysosomes for degradation. Here, we found that Ca2+ leakage after Mtb phagosome damage in human macrophages is the signal that triggers autophagy-related protein 8/microtubule-associated proteins 1A/1B light chain 3 (ATG8/LC3) lipidation. Unexpectedly, ATG8/LC3 lipidation did not target Mtb to lysosomes, excluding the canonical xenophagy. Upon Mtb phagosome damage, the Ca2+ leakage-dependent ATG8/LC3 lipidation occurred on multiple membranes instead of single or double membranes excluding the noncanonical autophagy pathways. Mechanistically, Ca2+ leakage from the phagosome triggered the recruitment of the V-ATPase-ATG16L1 complex independently of FIP200, ATG13, and proton gradient disruption. Furthermore, the Ca2+ leakage-dependent ATG8/LC3 lipidation limited Mtb phagosome damage and restricted Mtb replication. Together, we uncovered Ca2+ leakage as the key signal that triggers ATG8/LC3 lipidation on multiple membranes to mitigate Mtb phagosome damage.
Collapse
Affiliation(s)
- Di Chen
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Antony Fearns
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Maximiliano G. Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
8
|
Ben WB, Pirjo AM. ATG8 in single membranes: Fresh players of endocytosis and acidic organelle quality control in cancer, neurodegeneration, and inflammation. Biochem Biophys Res Commun 2025; 749:151384. [PMID: 39864381 DOI: 10.1016/j.bbrc.2025.151384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Ubiquitin-like autophagy-related gene ATG8 proteins are typically associated with degradative quality control via canonical double-membrane macro-autophagosomes in the cell. ATG8 proteins have now stepped forward in non-canonical pathways in single membrane organelles. The growing interest in non-canonical ATG8 roles has been stimulated by recent links to human conditions, especially in the regulation of inflammation, neurodegeneration and cancers. Here, we summarize the evidence linking non-canonical ATG8s to human pathologies and the quality control of acidic V-ATPase-regulated organelles in the cell.
Collapse
Affiliation(s)
- Wang B Ben
- Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
| | - Apaja M Pirjo
- Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia; College of Public Health and Medicine, Flinders University, Bedford Park, SA, 5042, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
| |
Collapse
|
9
|
Bentley-DeSousa A, Roczniak-Ferguson A, Ferguson SM. A STING-CASM-GABARAP pathway activates LRRK2 at lysosomes. J Cell Biol 2025; 224:e202310150. [PMID: 39812709 PMCID: PMC11734622 DOI: 10.1083/jcb.202310150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 09/28/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
Mutations that increase LRRK2 kinase activity have been linked to Parkinson's disease and Crohn's disease. LRRK2 is also activated by lysosome damage. However, the endogenous cellular mechanisms that control LRRK2 kinase activity are not well understood. In this study, we identify signaling through stimulator of interferon genes (STING) as an activator of LRRK2 via the conjugation of ATG8 to single membranes (CASM) pathway. We furthermore establish that multiple chemical stimuli that perturb lysosomal homeostasis also converge on CASM to activate LRRK2. Although CASM results in the lipidation of multiple ATG8 protein family members, we establish that LRRK2 lysosome recruitment and kinase activation are highly dependent on interactions with the GABARAP member of this family. Collectively, these results define a pathway that integrates multiple stimuli at lysosomes to control the kinase activity of LRRK2. Aberrant activation of LRRK2 via this pathway may be of relevance in both Parkinson's and Crohn's diseases.
Collapse
Affiliation(s)
- Amanda Bentley-DeSousa
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD, USA
| | - Agnes Roczniak-Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD, USA
| | - Shawn M. Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
10
|
Lorentzen KC, Prescott AR, Ganley IG. Artificial targeting of autophagy components to mitochondria reveals both conventional and unconventional mitophagy pathways. Autophagy 2025; 21:315-337. [PMID: 39177530 PMCID: PMC11760219 DOI: 10.1080/15548627.2024.2395149] [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: 04/07/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024] Open
Abstract
Macroautophagy/autophagy enables lysosomal degradation of a diverse array of intracellular material. This process is essential for normal cellular function and its dysregulation is implicated in many diseases. Given this, there is much interest in understanding autophagic mechanisms of action in order to determine how it can be best targeted therapeutically. In mitophagy, the selective degradation of mitochondria via autophagy, mitochondria first need to be primed with signals that allow the recruitment of the core autophagy machinery to drive the local formation of an autophagosome around the target mitochondrion. To determine how the recruitment of different core autophagy components can drive mitophagy, we took advantage of the mito-QC mitophagy assay (an outer mitochondrial membrane-localized tandem mCherry-GFP tag). By tagging autophagy proteins with an anti-mCherry (or anti-GFP) nanobody, we could recruit them to mitochondria and simultaneously monitor levels of mitophagy. We found that targeting ULK1, ATG16L1 and the different Atg8-family proteins was sufficient to induce mitophagy. Mitochondrial recruitment of ULK1 and the Atg8-family proteins induced a conventional mitophagy pathway, requiring RB1CC1/FIP200, PIK3C3/VPS34 activity and ATG5. Surprisingly, the mitophagy pathway upon recruitment of ATG16L1 proceeded independently of ATG5, although it still required RB1CC1 and PIK3C3/VPS34 activity. In this latter pathway, mitochondria were alternatively delivered to lysosomes via uptake into early endosomes.Abbreviation: aGFP: anti-GFP nanobody; amCh: anti-mCherry nanobody; ATG: autophagy related; ATG16L1: autophagy related 16 like 1; AUTAC/AUTOTAC: autophagy-targeting chimera; BafA1: bafilomycin A1; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CCCP: carbonyl cyanide m-chlorophenylhydrazone; COX4/COX IV: cytochrome c oxidase subunit 4; DFP: deferiprone; DMSO: dimethyl sulfoxide; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor associated protein like 1; HSPD1/HSP60: heat shock protein family D (Hsp60) member 1; HRP: horseradish peroxidase; HTRA2/OMI: HtrA serine peptidase 2; IB: immunoblotting; IF: immunofluorescence; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; NBR1: NBR1 autophagy cargo receptor; OMM: outer mitochondrial membrane; OPA1: OPA1 mitochondrial dynamin like GTPase; OPTN: optineurin; (D)PBS: (Dulbecco's) phosphate-buffered saline; PD: Parkinson disease; PFA: paraformaldehyde; POI: protein of interest; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; RAB: RAB, member RAS oncogene family; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SQSTM1: sequestosome 1; TAX1BP1: Tax1 binding protein 1; ULK: unc-51 like autophagy activating kinase 1; VPS: vacuolar protein sorting; WIPI: WD repeat domain, phosphoinositide interacting.
Collapse
Affiliation(s)
| | - Alan R. Prescott
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee, UK
| | - Ian G. Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| |
Collapse
|
11
|
Lee SK, Park SW, Jang DJ, Lee JA. Mechanisms and roles of membrane-anchored ATG8s. Front Cell Dev Biol 2025; 13:1532050. [PMID: 39936034 PMCID: PMC11810923 DOI: 10.3389/fcell.2025.1532050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Autophagy-related protein 8 (ATG8) family proteins, including LC3 and GABARAP subfamilies, are pivotal in canonical autophagy, driving autophagosome formation, cargo selection, and lysosomal fusion. However, recent studies have identified non-canonical roles for lipidated ATG8 in processes such as LC3-associated phagocytosis (LAP), LC3-associated endocytosis (LANDO), and lipidated ATG8-mediated secretory autophagy. These pathways expand ATG8's functional repertoire in immune regulation, membrane repair, and pathogen clearance, as ATG8 becomes conjugated to single-membrane structures (e.g., phagosomes and lysosomes). This review examines the molecular mechanisms of ATG8 lipidation, focusing on its selective conjugation to phosphatidylethanolamine (PE) in autophagy and phosphatidylserine (PS) in CASM. We highlight LIR-based probes and LC3/GABARAP-specific deconjugases as critical tools that allow precise tracking and manipulation of ATG8 in autophagic and non-autophagic contexts. These advancements hold therapeutic promise for treating autophagy-related diseases, including cancer and neurodegenerative disorders, by targeting ATG8-driven pathways that maintain cellular homeostasis.
Collapse
Affiliation(s)
- Soo-Kyeong Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Republic of Korea
| | - Sang-Won Park
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju, Republic of Korea
| | - Deok-Jin Jang
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju, Republic of Korea
| | - Jin-A. Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Republic of Korea
| |
Collapse
|
12
|
McMann E, Gorski SM. Last but not least: emerging roles of the autophagy-related protein ATG4D. Autophagy 2024; 20:1916-1927. [PMID: 38920354 PMCID: PMC11346562 DOI: 10.1080/15548627.2024.2369436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
The evolutionarily conserved ATG4 cysteine proteases regulate macroautophagy/autophagy through the priming and deconjugation of the Atg8-family proteins. In mammals there are four ATG4 family members (ATG4A, ATG4B, ATG4C, ATG4D) but ATG4D has been relatively understudied. Heightened interest in ATG4D has been stimulated by recent links to human disease. Notably, genetic variations in human ATG4D were implicated in a heritable neurodevelopmental disorder. Genetic analyses in dogs, along with loss-of-function zebrafish and mouse models, further support a neuroprotective role for ATG4D. Here we discuss the evidence connecting ATG4D to neurological diseases and other pathologies and summarize its roles in both autophagy-dependent and autophagy-independent cellular processes.Abbrevation: ATG: autophagy related; BafA1: bafilomycin A1; BCL2: BCL2 apoptosis regulator; BH3: BCL2 homology region 3; CASP3: caspase 3; EV: extracellular vesicle; GABA: gamma aminobutyric acid; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor associated protein like 1; GABARAPL2: GABA type A receptor associated protein like 2; GFP: green fluorescent protein; LIR: LC3-interacting region; MAP1LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; MYC: MYC proto-oncogene, bHLH transcription factor; PE: phosphatidylethanolamine; PS: phosphatidylserine; QKO: quadruple knockout; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel; SQSTM1: sequestosome 1.
Collapse
Affiliation(s)
- Emily McMann
- Canada’s Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Sharon M. Gorski
- Canada’s Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
13
|
Timimi L, Wrobel AG, Chiduza GN, Maslen SL, Torres-Méndez A, Montaner B, Davis C, Minckley T, Hole KL, Serio A, Devine MJ, Skehel JM, Rubinstein JL, Schreiber A, Beale R. The V-ATPase/ATG16L1 axis is controlled by the V 1H subunit. Mol Cell 2024; 84:2966-2983.e9. [PMID: 39089251 DOI: 10.1016/j.molcel.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 08/03/2024]
Abstract
Defects in organellar acidification indicate compromised or infected compartments. Recruitment of the autophagy-related ATG16L1 complex to pathologically neutralized organelles targets ubiquitin-like ATG8 molecules to perturbed membranes. How this process is coupled to proton gradient disruption is unclear. Here, we reveal that the V1H subunit of the vacuolar ATPase (V-ATPase) proton pump binds directly to ATG16L1. The V1H/ATG16L1 interaction only occurs within fully assembled V-ATPases, allowing ATG16L1 recruitment to be coupled to increased V-ATPase assembly following organelle neutralization. Cells lacking V1H fail to target ATG8s during influenza infection or after activation of the immune receptor stimulator of interferon genes (STING). We identify a loop within V1H that mediates ATG16L1 binding. A neuronal V1H isoform lacks this loop and is associated with attenuated ATG8 targeting in response to ionophores in primary murine and human iPSC-derived neurons. Thus, V1H controls ATG16L1 recruitment following proton gradient dissipation, suggesting that the V-ATPase acts as a cell-intrinsic damage sensor.
Collapse
Affiliation(s)
- Lewis Timimi
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Division of Medicine, University College London, London WC1E 6JF, UK
| | - Antoni G Wrobel
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - George N Chiduza
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Sarah L Maslen
- Proteomics STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Antonio Torres-Méndez
- Neural Circuits & Evolution Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Beatriz Montaner
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Colin Davis
- Cellular Degradation Systems Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Taylor Minckley
- Neural Circuit Bioengineering and Disease Modelling Laboratory, The Francis Crick Institute, London NW1 1AT, UK; UK Dementia Research Institute at King's College London, London SE5 9RX, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London SE5 9RX, UK
| | - Katriona L Hole
- Mitochondrial Neurobiology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Andrea Serio
- Neural Circuit Bioengineering and Disease Modelling Laboratory, The Francis Crick Institute, London NW1 1AT, UK; UK Dementia Research Institute at King's College London, London SE5 9RX, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London SE5 9RX, UK
| | - Michael J Devine
- Mitochondrial Neurobiology Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - J Mark Skehel
- Proteomics STP, The Francis Crick Institute, London NW1 1AT, UK
| | - John L Rubinstein
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, The University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medical Biophysics, The University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Anne Schreiber
- Cellular Degradation Systems Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Division of Medicine, University College London, London WC1E 6JF, UK.
| |
Collapse
|
14
|
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
|
15
|
Figueras-Novoa C, Timimi L, Marcassa E, Ulferts R, Beale R. Conjugation of ATG8s to single membranes at a glance. J Cell Sci 2024; 137:jcs261031. [PMID: 39145464 PMCID: PMC11361636 DOI: 10.1242/jcs.261031] [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: 08/16/2024] Open
Abstract
Autophagy refers to a set of degradative mechanisms whereby cytoplasmic contents are targeted to the lysosome. This is best described for macroautophagy, where a double-membrane compartment (autophagosome) is generated to engulf cytoplasmic contents. Autophagosomes are decorated with ubiquitin-like ATG8 molecules (ATG8s), which are recruited through covalent lipidation, catalysed by the E3-ligase-like ATG16L1 complex. LC3 proteins are ATG8 family members that are often used as a marker for autophagosomes. In contrast to canonical macroautophagy, conjugation of ATG8s to single membranes (CASM) describes a group of non-canonical autophagy processes in which ATG8s are targeted to pre-existing single-membrane compartments. CASM occurs in response to disrupted intracellular pH gradients, when the V-ATPase proton pump recruits ATG16L1 in a process called V-ATPase-ATG16L1-induced LC3 lipidation (VAIL). Recent work has demonstrated a parallel, alternative axis for CASM induction, triggered when the membrane recruitment factor TECPR1 recognises sphingomyelin exposed on the cytosolic face of a membrane and forms an alternative E3-ligase-like complex. This sphingomyelin-TECPR1-induced LC3 lipidation (STIL) is independent of the V-ATPase and ATG16L1. In light of these discoveries, this Cell Science at a Glance article summarises these two mechanisms of CASM to highlight how they differ from canonical macroautophagy, and from each other.
Collapse
Affiliation(s)
- Carmen Figueras-Novoa
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Lewis Timimi
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
- Division of Medicine, University College London, London NW1 1AT, UK
| | - Elena Marcassa
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Rachel Ulferts
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
- Division of Medicine, University College London, London NW1 1AT, UK
| |
Collapse
|
16
|
Knyazeva A, Li S, Corkery DP, Shankar K, Herzog LK, Zhang X, Singh B, Niggemeyer G, Grill D, Gilthorpe JD, Gaetani M, Carlson LA, Waldmann H, Wu YW. A chemical inhibitor of IST1-CHMP1B interaction impairs endosomal recycling and induces noncanonical LC3 lipidation. Proc Natl Acad Sci U S A 2024; 121:e2317680121. [PMID: 38635626 PMCID: PMC11047075 DOI: 10.1073/pnas.2317680121] [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: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 04/20/2024] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) machinery constitutes multisubunit protein complexes that play an essential role in membrane remodeling and trafficking. ESCRTs regulate a wide array of cellular processes, including cytokinetic abscission, cargo sorting into multivesicular bodies (MVBs), membrane repair, and autophagy. Given the versatile functionality of ESCRTs, and the intricate organizational structure of the ESCRT machinery, the targeted modulation of distinct ESCRT complexes is considerably challenging. This study presents a pseudonatural product targeting IST1-CHMP1B within the ESCRT-III complexes. The compound specifically disrupts the interaction between IST1 and CHMP1B, thereby inhibiting the formation of IST1-CHMP1B copolymers essential for normal-topology membrane scission events. While the compound has no impact on cytokinesis, MVB sorting, or biogenesis of extracellular vesicles, it rapidly inhibits transferrin receptor recycling in cells, resulting in the accumulation of transferrin in stalled sorting endosomes. Stalled endosomes become decorated by lipidated LC3, suggesting a link between noncanonical LC3 lipidation and inhibition of the IST1-CHMP1B complex.
Collapse
Affiliation(s)
- Anastasia Knyazeva
- Department of Chemistry, Umeå University, 901 87Umeå, Sweden
- Science for Life Laboratory, Umeå University, 901 87Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 901 87Umeå, Sweden
| | - Shuang Li
- Department of Chemistry, Umeå University, 901 87Umeå, Sweden
- Science for Life Laboratory, Umeå University, 901 87Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 901 87Umeå, Sweden
| | - Dale P. Corkery
- Department of Chemistry, Umeå University, 901 87Umeå, Sweden
- Science for Life Laboratory, Umeå University, 901 87Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 901 87Umeå, Sweden
| | - Kasturika Shankar
- Umeå Centre for Microbial Research, Umeå University, 901 87Umeå, Sweden
- Department of Medical Biochemistry and Biophysics, 901 87Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden
- Molecular Infection Medicine Sweden, Umeå University, 901 87, Umeå, Sweden
| | - Laura K. Herzog
- Department of Chemistry, Umeå University, 901 87Umeå, Sweden
- Science for Life Laboratory, Umeå University, 901 87Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 901 87Umeå, Sweden
| | - Xuepei Zhang
- Chemical Proteomics Core Facility, Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77Stockholm, Sweden
- Chemical Proteomics Unit, Science for Life Laboratory, 171 77Stockholm, Sweden
- Chemical Proteomics, Swedish National Infrastructure for Biological Mass Spectrometry, 171 77Stockholm, Sweden
| | - Birendra Singh
- Department of Surgical and Perioperative Sciences, Unit of Anesthesiology and Intensive Care Medicine, Umeå University, 901 87Umeå, Sweden
| | - Georg Niggemeyer
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - David Grill
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | | | - Massimiliano Gaetani
- Chemical Proteomics Core Facility, Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77Stockholm, Sweden
- Chemical Proteomics Unit, Science for Life Laboratory, 171 77Stockholm, Sweden
- Chemical Proteomics, Swedish National Infrastructure for Biological Mass Spectrometry, 171 77Stockholm, Sweden
| | - Lars-Anders Carlson
- Umeå Centre for Microbial Research, Umeå University, 901 87Umeå, Sweden
- Department of Medical Biochemistry and Biophysics, 901 87Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden
- Molecular Infection Medicine Sweden, Umeå University, 901 87, Umeå, Sweden
| | - Herbert Waldmann
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
- Faculty of Chemistry and Chemical Biology, Technical University Dortmund, 44227, Dortmund, Germany
| | - Yao-Wen Wu
- Department of Chemistry, Umeå University, 901 87Umeå, Sweden
- Science for Life Laboratory, Umeå University, 901 87Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 901 87Umeå, Sweden
| |
Collapse
|
17
|
Tang J, Fang D, Zhong J, Li M. Missing WD40 Repeats in ATG16L1 Delays Canonical Autophagy and Inhibits Noncanonical Autophagy. Int J Mol Sci 2024; 25:4493. [PMID: 38674078 PMCID: PMC11050548 DOI: 10.3390/ijms25084493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Canonical autophagy is an evolutionarily conserved process that forms double-membrane structures and mediates the degradation of long-lived proteins (LLPs). Noncanonical autophagy (NCA) is an important alternative pathway involving the formation of microtubule-associated protein 1 light chain 3 (LC3)-positive structures that are independent of partial core autophagy proteins. NCA has been defined by the conjugation of ATG8s to single membranes (CASM). During canonical autophagy and NCA/CASM, LC3 undergoes a lipidation modification, and ATG16L1 is a crucial protein in this process. Previous studies have reported that the WDR domain of ATG16L1 is not necessary for canonical autophagy. However, our study found that WDR domain deficiency significantly impaired LLP degradation in basal conditions and slowed down LC3-II accumulation in canonical autophagy. We further demonstrated that the observed effect was due to a reduced interaction between ATG16L1 and FIP200/WIPI2, without affecting lysosome function or fusion. Furthermore, we also found that the WDR domain of ATG16L1 is crucial for chemical-induced NCA/CASM. The results showed that removing the WDR domain or introducing the K490A mutation in ATG16L1 significantly inhibited the NCA/CASM, which interrupted the V-ATPase-ATG16L1 axis. In conclusion, this study highlights the significance of the WDR domain of ATG16L1 for both canonical autophagy and NCA functions, improving our understanding of its role in autophagy.
Collapse
Affiliation(s)
- Jiuge Tang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangzhou 510006, China
| | - Dongmei Fang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangzhou 510006, China
| | - Jialing Zhong
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangzhou 510006, China
| | - Min Li
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangzhou 510006, China
| |
Collapse
|
18
|
Eguchi T, Sakurai M, Wang Y, Saito C, Yoshii G, Wileman T, Mizushima N, Kuwahara T, Iwatsubo T. The V-ATPase-ATG16L1 axis recruits LRRK2 to facilitate the lysosomal stress response. J Cell Biol 2024; 223:e202302067. [PMID: 38227290 PMCID: PMC10791558 DOI: 10.1083/jcb.202302067] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 10/13/2023] [Accepted: 12/11/2023] [Indexed: 01/17/2024] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2), a Rab kinase associated with Parkinson's disease and several inflammatory diseases, has been shown to localize to stressed lysosomes and get activated to regulate lysosomal homeostasis. However, the mechanisms of LRRK2 recruitment and activation have not been well understood. Here, we found that the ATG8 conjugation system regulates the recruitment of LRRK2 as well as LC3 onto single membranes of stressed lysosomes/phagosomes. This recruitment did not require FIP200-containing autophagy initiation complex, nor did it occur on double-membrane autophagosomes, suggesting independence from canonical autophagy. Consistently, LRRK2 recruitment was regulated by the V-ATPase-ATG16L1 axis, which requires the WD40 domain of ATG16L1 and specifically mediates ATG8 lipidation on single membranes. This mechanism was also responsible for the lysosomal stress-induced activation of LRRK2 and the resultant regulation of lysosomal secretion and enlargement. These results indicate that the V-ATPase-ATG16L1 axis serves a novel non-autophagic role in the maintenance of lysosomal homeostasis by recruiting LRRK2.
Collapse
Affiliation(s)
- Tomoya Eguchi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Maria Sakurai
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yingxue Wang
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Chieko Saito
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Gen Yoshii
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoki Kuwahara
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Chang C, You H, Su H, Hung I, Kao C, Huang S. Anti-influenza A (H1N1) virus effect of gallic acid through inhibition of virulent protein production and association with autophagy. Food Sci Nutr 2024; 12:1605-1615. [PMID: 38455214 PMCID: PMC10916620 DOI: 10.1002/fsn3.3852] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 03/09/2024] Open
Abstract
Influenza remains one of the most serious infectious diseases. Gallic acid is one of the most common and representative phenolic acids found in various plants. This is an interesting subject to explore how gallic acid could inhibit H1N1 influenza virus infection by reducing the production of virulent proteins and interrupting autophagy machinery for influenza virus replication on the host cell. Cellular viability was assessed by XTT assay. The inhibitory effects on the H1N1 influenza virus were assessed by hemagglutination assay, plaque assay, and qRT-PCR. Western blot analysis was used for detecting protein levels of M1, M2, NP, LC3B, and beclin-1. Autophagy activity was demonstrated by acridine orange staining assay. The result demonstrated that there was no cytotoxic effect of gallic acid on A549 cells, and gallic acid could restore the cellular viability of H1N1 influenza virus-infected A549 cells within the experimental concentration treatment. Moreover, gallic acid could effectively restrain viral activity of the H1N1 influenza virus. After the treatment of gallic acid, the production of virulent H1N1 influenza virus proteins, that is, M1, M2, and NP protein were reduced. As for autophagic mechanism, both of the LC3B II conversion and the level ratio of LC3B II to LC3B I were notably decreased. The acridine orange staining assay also revealed decreased accumulation of autophagosomes in H1N1 influenza virus-infected cells. In conclusion, gallic acid suppresses H1N1 influenza viral infectivity through restoration of autophagy pathway and inhibition of virulent M1, M2, and NP protein production.
Collapse
Affiliation(s)
- Cheng‐Chieh Chang
- Department of Chinese MedicineKaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiungTaiwan
- Graduate Institute of Chinese MedicineChina Medical UniversityTaichungTaiwan
| | - Huey‐Ling You
- Department of Laboratory MedicineKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
| | - Huey‐Jen Su
- Department of NursingMeiho UniversityNeipu ShiangTaiwan
| | - I‐Ling Hung
- Department of Chinese MedicineKaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiungTaiwan
- Department of Chinese MedicineJen‐Ai HospitalTaichungTaiwan
| | - Chao‐Wei Kao
- Department of Chinese MedicineKaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiungTaiwan
| | - Sheng‐Teng Huang
- Department of Chinese MedicineChina Medical University HospitalTaichungTaiwan
- School of Chinese MedicineChina Medical UniversityTaichungTaiwan
- An‐Nan HospitalChina Medical UniversityTainanTaiwan
- Cancer Research Center for Traditional Chinese Medicine, Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
| |
Collapse
|
20
|
Liu B. Protons are the new first responders. Science 2024; 383:713-714. [PMID: 38359124 DOI: 10.1126/science.adn9407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Human STING's newfound function as a channel expands our understanding of immunity.
Collapse
Affiliation(s)
- Bingxu Liu
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| |
Collapse
|
21
|
Cross J, Durgan J, McEwan DG, Tayler M, Ryan KM, Florey O. Lysosome damage triggers direct ATG8 conjugation and ATG2 engagement via non-canonical autophagy. J Cell Biol 2023; 222:e202303078. [PMID: 37796195 PMCID: PMC10561555 DOI: 10.1083/jcb.202303078] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/14/2023] [Accepted: 09/07/2023] [Indexed: 10/06/2023] Open
Abstract
Cells harness multiple pathways to maintain lysosome integrity, a central homeostatic process. Damaged lysosomes can be repaired or targeted for degradation by lysophagy, a selective autophagy process involving ATG8/LC3. Here, we describe a parallel ATG8/LC3 response to lysosome damage, mechanistically distinct from lysophagy. Using a comprehensive series of biochemical, pharmacological, and genetic approaches, we show that lysosome damage induces non-canonical autophagy and Conjugation of ATG8s to Single Membranes (CASM). Following damage, ATG8s are rapidly and directly conjugated onto lysosome membranes, independently of ATG13/WIPI2, lipidating to PS (and PE), a molecular hallmark of CASM. Lysosome damage drives V-ATPase V0-V1 association, direct recruitment of ATG16L1 via its WD40-domain/K490A, and is sensitive to Salmonella SopF. Lysosome damage-induced CASM is associated with formation of dynamic, LC3A-positive tubules, and promotes robust LC3A engagement with ATG2, a lipid transfer protein central to lysosome repair. Together, our data identify direct ATG8 conjugation as a rapid response to lysosome damage, with important links to lipid transfer and dynamics.
Collapse
Affiliation(s)
- Jake Cross
- Signalling Programme, Babraham Institute, Cambridge, UK
| | - Joanne Durgan
- Signalling Programme, Babraham Institute, Cambridge, UK
| | - David G. McEwan
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Kevin M. Ryan
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, UK
| | - Oliver Florey
- Signalling Programme, Babraham Institute, Cambridge, UK
| |
Collapse
|
22
|
Sun Y, Wang X, Yang X, Wang L, Ding J, Wang CC, Zhang H, Wang X. V-ATPase recruitment to ER exit sites switches COPII-mediated transport to lysosomal degradation. Dev Cell 2023; 58:2761-2775.e5. [PMID: 37922908 DOI: 10.1016/j.devcel.2023.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 08/23/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
Endoplasmic reticulum (ER)-phagy is crucial to regulate the function and homeostasis of the ER via lysosomal degradation, but how it is initiated is unclear. Here we discover that Z-AAT, a disease-causing mutant of α1-antitrypsin, induces noncanonical ER-phagy at ER exit sites (ERESs). Accumulation of misfolded Z-AAT at the ERESs impairs coat protein complex II (COPII)-mediated ER-to-Golgi transport and retains V0 subunits that further assemble V-ATPase at the arrested ERESs. V-ATPase subsequently recruits ATG16L1 onto ERESs to mediate in situ lipidation of LC3C. FAM134B-II is then recruited by LC3C via its LIR motif and elicits ER-phagy leading to efficient lysosomal degradation of Z-AAT. Activation of this ER-phagy mediated by the V-ATPase-ATG16L1-LC3C axis (EVAC) is also triggered by blocking ER export. Our findings identify a pathway which switches COPII-mediated transport to lysosomal degradation for ER quality control.
Collapse
Affiliation(s)
- Yiwei Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi'e Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaotong Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingjin Ding
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chih-Chen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
23
|
Florey O. TECPR1 helps bridge the CASM during lysosome damage. EMBO J 2023; 42:e115210. [PMID: 37638605 PMCID: PMC10548167 DOI: 10.15252/embj.2023115210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023] Open
Abstract
Maintaining the integrity of the endolysosomal system is of great importance for cellular homeostasis. Recent work published in The EMBO Journal and EMBO Reports reveals a novel role for the protein TECPR1 as a sensor for stressed membranes and regulator of lysosomal membrane repair.
Collapse
|
24
|
Deretic V. Atg8ylation as a host-protective mechanism against Mycobacterium tuberculosis. FRONTIERS IN TUBERCULOSIS 2023; 1:1275882. [PMID: 37901138 PMCID: PMC10612523 DOI: 10.3389/ftubr.2023.1275882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Nearly two decades have passed since the first report on autophagy acting as a cell-autonomous defense against Mycobacterium tuberculosis. This helped usher a new area of research within the field of host-pathogen interactions and led to the recognition of autophagy as an immunological mechanism. Interest grew in the fundamental mechanisms of antimicrobial autophagy and in the prophylactic and therapeutic potential for tuberculosis. However, puzzling in vivo data have begun to emerge in murine models of M. tuberculosis infection. The control of infection in mice affirmed the effects of certain autophagy genes, specifically ATG5, but not of other ATGs. Recent studies with a more complete inactivation of ATG genes now show that multiple ATG genes are indeed necessary for protection against M. tuberculosis. These particular ATG genes are involved in the process of membrane atg8ylation. Atg8ylation in mammalian cells is a broad response to membrane stress, damage and remodeling of which canonical autophagy is one of the multiple downstream outputs. The current developments clarify the controversies and open new avenues for both fundamental and translational studies.
Collapse
Affiliation(s)
- 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
| |
Collapse
|
25
|
Boyle KB, Ellison CJ, Elliott PR, Schuschnig M, Grimes K, Dionne MS, Sasakawa C, Munro S, Martens S, Randow F. TECPR1 conjugates LC3 to damaged endomembranes upon detection of sphingomyelin exposure. EMBO J 2023; 42:e113012. [PMID: 37409490 PMCID: PMC10476172 DOI: 10.15252/embj.2022113012] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023] Open
Abstract
Invasive bacteria enter the cytosol of host cells through initial uptake into bacteria-containing vacuoles (BCVs) and subsequent rupture of the BCV membrane, thereby exposing to the cytosol intraluminal, otherwise shielded danger signals such as glycans and sphingomyelin. The detection of glycans by galectin-8 triggers anti-bacterial autophagy, but how cells sense and respond to cytosolically exposed sphingomyelin remains unknown. Here, we identify TECPR1 (tectonin beta-propeller repeat containing 1) as a receptor for cytosolically exposed sphingomyelin, which recruits ATG5 into an E3 ligase complex that mediates lipid conjugation of LC3 independently of ATG16L1. TECPR1 binds sphingomyelin through its N-terminal DysF domain (N'DysF), a feature not shared by other mammalian DysF domains. Solving the crystal structure of N'DysF, we identified key residues required for the interaction, including a solvent-exposed tryptophan (W154) essential for binding to sphingomyelin-positive membranes and the conjugation of LC3 to lipids. Specificity of the ATG5/ATG12-E3 ligase responsible for the conjugation of LC3 is therefore conferred by interchangeable receptor subunits, that is, the canonical ATG16L1 and the sphingomyelin-specific TECPR1, in an arrangement reminiscent of certain multi-subunit ubiquitin E3 ligases.
Collapse
Affiliation(s)
- Keith B Boyle
- Division of Protein and Nucleic Acid ChemistryMRC Laboratory of Molecular BiologyCambridgeUK
| | - Cara J Ellison
- Division of Protein and Nucleic Acid ChemistryMRC Laboratory of Molecular BiologyCambridgeUK
- Present address:
Department of BiochemistryUniversity of OxfordOxfordUK
| | - Paul R Elliott
- Division of Protein and Nucleic Acid ChemistryMRC Laboratory of Molecular BiologyCambridgeUK
- Present address:
Department of BiochemistryUniversity of OxfordOxfordUK
| | - Martina Schuschnig
- Max Perutz Labs, Vienna BioCenter (VBC)University of ViennaViennaAustria
| | - Krista Grimes
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUK
| | - Marc S Dionne
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUK
| | - Chihiro Sasakawa
- Medical Mycology Research CenterChiba UniversityChibaJapan
- Nippon Institute for Biological ScienceOmeJapan
| | - Sean Munro
- Division of Protein and Nucleic Acid ChemistryMRC Laboratory of Molecular BiologyCambridgeUK
| | - Sascha Martens
- Max Perutz Labs, Vienna BioCenter (VBC)University of ViennaViennaAustria
- Center for Molecular Biology, Department of Biochemistry and Cell BiologyUniversity of ViennaViennaAustria
| | - Felix Randow
- Division of Protein and Nucleic Acid ChemistryMRC Laboratory of Molecular BiologyCambridgeUK
- Department of Medicine, Addenbrooke's HospitalUniversity of CambridgeCambridgeUK
| |
Collapse
|
26
|
Liu B, Carlson RJ, Pires IS, Gentili M, Feng E, Hellier Q, Schwartz MA, Blainey PC, Irvine DJ, Hacohen N. Human STING is a proton channel. Science 2023; 381:508-514. [PMID: 37535724 PMCID: PMC11260435 DOI: 10.1126/science.adf8974] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 06/30/2023] [Indexed: 08/05/2023]
Abstract
Proton leakage from organelles is a common signal for noncanonical light chain 3B (LC3B) lipidation and inflammasome activation, processes induced upon stimulator of interferon genes (STING) activation. On the basis of structural analysis, we hypothesized that human STING is a proton channel. Indeed, we found that STING activation induced a pH increase in the Golgi and that STING reconstituted in liposomes enabled transmembrane proton transport. Compound 53 (C53), a STING agonist that binds the putative channel interface, blocked STING-induced proton flux in the Golgi and in liposomes. STING-induced LC3B lipidation and inflammasome activation were also inhibited by C53, suggesting that STING's channel activity is critical for these two processes. Thus, STING's interferon-induction function can be decoupled from its roles in LC3B lipidation and inflammasome activation.
Collapse
Affiliation(s)
- Bingxu Liu
- Broad Institute, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
| | - Rebecca J. Carlson
- Broad Institute, Cambridge, MA, USA
- Massachusetts Institute of Technology, Department of Health Sciences and Technology, Cambridge, MA, USA
| | - Ivan S. Pires
- The Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
| | | | - Ellie Feng
- Broad Institute, Cambridge, MA, USA
- Massachusetts Institute of Technology, Department of Biological Engineering, Cambridge, MA, USA
| | | | - Marc A. Schwartz
- Broad Institute, Cambridge, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Hematology and Oncology, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paul C. Blainey
- Broad Institute, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
- Massachusetts Institute of Technology, Department of Biological Engineering, Cambridge, MA, USA
| | - Darrell J. Irvine
- The Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
| | - Nir Hacohen
- Broad Institute, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
27
|
Javed R, Jain A, Duque T, Hendrix E, Paddar MA, Khan S, Claude‐Taupin A, Jia J, Allers L, Wang F, Mudd M, Timmins G, Lidke K, Rusten TE, Akepati PR, He Y, Reggiori F, Eskelinen E, Deretic V. Mammalian ATG8 proteins maintain autophagosomal membrane integrity through ESCRTs. EMBO J 2023; 42:e112845. [PMID: 37272163 PMCID: PMC10350836 DOI: 10.15252/embj.2022112845] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
The canonical autophagy pathway in mammalian cells sequesters diverse cytoplasmic cargo within the double membrane autophagosomes that eventually convert into degradative compartments via fusion with endolysosomal intermediates. Here, we report that autophagosomal membranes show permeability in cells lacking principal ATG8 proteins (mATG8s) and are unable to mature into autolysosomes. Using a combination of methods including a novel in vitro assay to measure membrane sealing, we uncovered a previously unappreciated function of mATG8s to maintain autophagosomal membranes in a sealed state. The mATG8 proteins GABARAP and LC3A bind to key ESCRT-I components contributing, along with other ESCRTs, to the integrity and imperviousness of autophagic membranes. Autophagic organelles in cells lacking mATG8s are permeant, are arrested as amphisomes, and do not progress to functional autolysosomes. Thus, autophagosomal organelles need to be maintained in a sealed state in order to become lytic autolysosomes.
Collapse
Affiliation(s)
- Ruheena Javed
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Ashish Jain
- Faculty of MedicineUniversity of OsloOsloNorway
| | - Thabata Duque
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Emily Hendrix
- Department of Chemistry & Chemical BiologyThe University of New MexicoAlbuquerqueNMUSA
| | - Masroor Ahmad Paddar
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Sajjad Khan
- Department of Physics and AstronomyThe University of New MexicoAlbuquerqueNMUSA
| | - Aurore Claude‐Taupin
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Jingyue Jia
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Lee Allers
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Fulong Wang
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Michal Mudd
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Graham Timmins
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Keith Lidke
- Department of Physics and AstronomyThe University of New MexicoAlbuquerqueNMUSA
| | | | - Prithvi Reddy Akepati
- Division of Gastroenterology and Hepatology, Department of Internal MedicineUniversity of New MexicoAlbuquerqueNMUSA
| | - Yi He
- Department of Chemistry & Chemical BiologyThe University of New MexicoAlbuquerqueNMUSA
| | - Fulvio Reggiori
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Aarhus Institute for Advanced Studies (AIAS)Aarhus UniversityAarhusDenmark
| | | | - Vojo Deretic
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| |
Collapse
|
28
|
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
|
29
|
Abstract
Macroautophagy and microautophagy are highly conserved eukaryotic cellular processes that degrade cytoplasmic material in lysosomes. Both pathways involve characteristic membrane dynamics regulated by autophagy-related proteins and other molecules, some of which are shared between the two pathways. Over the past few years, the application of new technologies, such as cryo-electron microscopy, coevolution-based structural prediction and in vitro reconstitution, has revealed the functions of individual autophagy gene products, especially in autophagy induction, membrane reorganization and cargo recognition. Concomitantly, mutations in autophagy genes have been linked to human disorders, particularly neurodegenerative diseases, emphasizing the potential pathogenic implications of autophagy defects. Accumulating genome data have also illuminated the evolution of autophagy genes within eukaryotes as well as their transition from possible ancestral elements in prokaryotes.
Collapse
Affiliation(s)
- Hayashi Yamamoto
- grid.26999.3d0000 0001 2151 536XDepartment of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan ,grid.410821.e0000 0001 2173 8328Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Sidi Zhang
- grid.26999.3d0000 0001 2151 536XDepartment of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noboru Mizushima
- grid.26999.3d0000 0001 2151 536XDepartment of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
Serramito-Gómez I, Terraza-Silvestre E, Fernández-Cabrera Á, Villamuera R, Pimentel-Muiños FX. ATG16L1 WD40 domain-dependent IL10R (interleukin 10 receptor) signaling is insensitive to the T300A Crohn disease risk polymorphism. Autophagy 2022; 18:3023-3030. [PMID: 35311452 PMCID: PMC9673950 DOI: 10.1080/15548627.2022.2054241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A coding allele of ATG16L1 that increases the risk of Crohn disease (T300A; rs2241880) impairs the interaction between the C-terminal WD40 domain (WDD) and proteins containing a WDD-binding motif, thus specifically inhibiting the unconventional autophagic activities of ATG16L1. In a recent publication we described a novel atypical role of ATG16L1 in the regulation of IL10R (interleukin 10 receptor) trafficking and signaling, an activity that involves direct interaction between the WDD and a target motif present in IL10RB (interleukin 10 receptor subunit beta). Here we show that, unexpectedly, neither the ability of ATG16L1 to interact with IL10RB nor its role in supporting IL10 signaling are altered by the T300A mutation. These results indicate that the ATG16L1T300A allele selectively impairs the interaction between the WDD and a subset of WDD-binding motif versions, suggesting that only a fraction of the unconventional activities mediated by ATG16L1 are required to prevent Crohn disease.Abbreviations: ATG, autophagy related; ATG16L1, autophagy related 16 like 1; BMDMs, bone marrow-derived macrophages; CRISPR, clustered regularly interspaced short palindromic repeats; CSF1/M-CSF, colony stimulating factor 1; FBS, fetal bovine serum; GSH, glutathione; IL10, interleukin 10; IL10R, interleukin 10 receptor; LPS, lipopolysaccharide; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; MEFs, mouse embryonic fibroblasts; PMA, phorbol myristate acetate; p-STAT3: phosphorylated STAT3; qPCR, quantitative polymerase chain reaction; SDS, sodium dodecyl sulfate; sgRNA, single guide RNA; TMEM59, transmembrane protein 59; TNF, tumor necrosis factor; TNFAIP3/A20, TNF alpha induced protein 3; WDD, WD40 domain; WIPI2, WD repeat domain, phosphoinositide interacting 2.
Collapse
Affiliation(s)
- Inmaculada Serramito-Gómez
- Instituto de Biología Molecular y Celular del CáncerCentro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Elena Terraza-Silvestre
- Instituto de Biología Molecular y Celular del CáncerCentro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Álvaro Fernández-Cabrera
- Instituto de Biología Molecular y Celular del CáncerCentro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Villamuera
- Instituto de Biología Molecular y Celular del CáncerCentro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Felipe X. Pimentel-Muiños
- Instituto de Biología Molecular y Celular del CáncerCentro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain,CONTACT Felipe X. Pimentel-Muiños ;Instituto de Biología Molecular y Celular del Cáncer. Centro de Investigación del Cáncer, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera, 1. Salamanca, Madrid, 28049, Spain
| |
Collapse
|
31
|
Wang Y, Ramos M, Jefferson M, Zhang W, Beraza N, Carding S, Powell PP, Stewart JP, Mayer U, Wileman T. Control of infection by LC3-associated phagocytosis, CASM, and detection of raised vacuolar pH by the V-ATPase-ATG16L1 axis. SCIENCE ADVANCES 2022; 8:eabn3298. [PMID: 36288298 PMCID: PMC9604538 DOI: 10.1126/sciadv.abn3298] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 07/07/2022] [Indexed: 05/29/2023]
Abstract
The delivery of pathogens to lysosomes for degradation provides an important defense against infection. Degradation is enhanced when LC3 is conjugated to endosomes and phagosomes containing pathogens to facilitate fusion with lysosomes. In phagocytic cells, TLR signaling and Rubicon activate LC3-associated phagocytosis (LAP) where stabilization of the NADPH oxidase leads to sustained ROS production and raised vacuolar pH. Raised pH triggers the assembly of the vacuolar ATPase on the vacuole membrane where it binds ATG16L1 to recruit the core LC3 conjugation complex (ATG16L1:ATG5-12). This V-ATPase-ATG16L1 axis is also activated in nonphagocytic cells to conjugate LC3 to endosomes containing extracellular microbes. Pathogens provide additional signals for recruitment of LC3 when they raise vacuolar pH with pore-forming toxins and proteins, phospholipases, or specialized secretion systems. Many microbes secrete virulence factors to inhibit ROS production and/or the V-ATPase-ATG16L1 axis to slow LC3 recruitment and avoid degradation in lysosomes.
Collapse
Affiliation(s)
- Yingxue Wang
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| | - Maria Ramos
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| | | | - Weijiao Zhang
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | | | - Penny P. Powell
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - James P. Stewart
- Department of Infection Biology, University of Liverpool, Liverpool, UK
| | - Ulrike Mayer
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| |
Collapse
|
32
|
Durgan J, Florey O. Many roads lead to CASM: Diverse stimuli of noncanonical autophagy share a unifying molecular mechanism. SCIENCE ADVANCES 2022; 8:eabo1274. [PMID: 36288315 PMCID: PMC9604613 DOI: 10.1126/sciadv.abo1274] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Autophagy is a fundamental catabolic process coordinated by a network of autophagy-related (ATG) proteins. These ATG proteins also perform an important parallel role in "noncanonical" autophagy, a lysosome-associated signaling pathway with key functions in immunity, inflammation, cancer, and neurodegeneration. While the noncanonical autophagy pathway shares the common ATG machinery, it bears key mechanistic and functional distinctions, and is characterized by conjugation of ATG8 to single membranes (CASM). Here, we review the diverse, and still expanding, collection of stimuli and processes now known to harness the noncanonical autophagy pathway, including engulfment processes, drug treatments, TRPML1 and STING signaling, viral infection, and other pathogenic factors. We discuss the multiple associated routes to CASM and assess their shared and distinctive molecular features. By integrating these findings, we propose an updated and unifying mechanism for noncanonical autophagy, centered on ATG16L1 and V-ATPase.
Collapse
|
33
|
Magné J, Green DR. LC3-associated endocytosis and the functions of Rubicon and ATG16L1. SCIENCE ADVANCES 2022; 8:eabo5600. [PMID: 36288306 PMCID: PMC9604520 DOI: 10.1126/sciadv.abo5600] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
LC3-associated endocytosis (LANDO) is a noncanonical function of the autophagy machinery, in which LC3 (microtubule-associated protein light chain) is conjugated to rab5-positive endosomes, using a portion of the canonical autophagy pathway. LANDO was initially discovered in a murine model of Alzheimer's disease as a critical regulator of amyloid-β receptor recycling in microglial cells, playing a protective role against neuronal loss and memory impairment. Recent evidence suggests an emerging role of LANDO in cytokine receptor signaling and innate immunity. Here, we discuss the regulation of two crucial effectors of LANDO, Rubicon and ATG16L1, and their impact on endocytosis, autophagy, and phagocytosis.
Collapse
|
34
|
Pant A, Yao X, Lavedrine A, Viret C, Dockterman J, Chauhan S, Chong-Shan Shi, Manjithaya R, Cadwell K, Kufer TA, Kehrl JH, Coers J, Sibley LD, Faure M, Taylor GA, Chauhan S. Interactions of Autophagy and the Immune System in Health and Diseases. AUTOPHAGY REPORTS 2022; 1:438-515. [PMID: 37425656 PMCID: PMC10327624 DOI: 10.1080/27694127.2022.2119743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Autophagy is a highly conserved process that utilizes lysosomes to selectively degrade a variety of intracellular cargo, thus providing quality control over cellular components and maintaining cellular regulatory functions. Autophagy is triggered by multiple stimuli ranging from nutrient starvation to microbial infection. Autophagy extensively shapes and modulates the inflammatory response, the concerted action of immune cells, and secreted mediators aimed to eradicate a microbial infection or to heal sterile tissue damage. Here, we first review how autophagy affects innate immune signaling, cell-autonomous immune defense, and adaptive immunity. Then, we discuss the role of non-canonical autophagy in microbial infections and inflammation. Finally, we review how crosstalk between autophagy and inflammation influences infectious, metabolic, and autoimmune disorders.
Collapse
Affiliation(s)
- Aarti Pant
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Xiaomin Yao
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Aude Lavedrine
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM
| | - Jake Dockterman
- Department of Immunology, Duke University, Medical Center, Durham, North Carolina, USA
| | - Swati Chauhan
- Cell biology and Infectious diseases, Institute of Life Sciences, Bhubaneswar, India
| | - Chong-Shan Shi
- Laboratory of Immunoregulation, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, United States of America
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Thomas A. Kufer
- Department of Immunology, Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - John H. Kehrl
- Laboratory of Immunoregulation, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jörn Coers
- Department of Immunology, Duke University, Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Medical Center, Durham, North Carolina, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University Sch. Med., St Louis, MO, 63110, USA
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM
| | - Gregory A Taylor
- Department of Immunology, Duke University, Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Medical Center, Durham, North Carolina, USA
- Department of Molecular Microbiology, Washington University Sch. Med., St Louis, MO, 63110, USA
- Geriatric Research, Education, and Clinical Center, VA Health Care Center, Durham, North Carolina, USA
- Departments of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development, Duke University, Medical Center, Durham, North Carolina, USA
| | - Santosh Chauhan
- Cell biology and Infectious diseases, Institute of Life Sciences, Bhubaneswar, India
- CSIR–Centre For Cellular And Molecular Biology (CCMB), Hyderabad, Telangana
| |
Collapse
|
35
|
Jia X, Knyazeva A, Zhang Y, Castro-Gonzalez S, Nakamura S, Carlson LA, Yoshimori T, Corkery DP, Wu YW. V. cholerae MakA is a cholesterol-binding pore-forming toxin that induces non-canonical autophagy. J Cell Biol 2022; 221:213518. [PMID: 36194176 PMCID: PMC9536202 DOI: 10.1083/jcb.202206040] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/16/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023] Open
Abstract
Pore-forming toxins (PFTs) are important virulence factors produced by many pathogenic bacteria. Here, we show that the Vibrio cholerae toxin MakA is a novel cholesterol-binding PFT that induces non-canonical autophagy in a pH-dependent manner. MakA specifically binds to cholesterol on the membrane at pH < 7. Cholesterol-binding leads to oligomerization of MakA on the membrane and pore formation at pH 5.5. Unlike other cholesterol-dependent cytolysins (CDCs) which bind cholesterol through a conserved cholesterol-binding motif (Thr-Leu pair), MakA contains an Ile-Ile pair that is essential for MakA-cholesterol interaction. Following internalization, endosomal acidification triggers MakA pore-assembly followed by ESCRT-mediated membrane repair and V-ATPase-dependent unconventional LC3 lipidation on the damaged endolysosomal membranes. These findings characterize a new cholesterol-binding toxin that forms pores in a pH-dependent manner and reveals the molecular mechanism of host autophagy manipulation.
Collapse
Affiliation(s)
- Xiaotong Jia
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Anastasia Knyazeva
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Yu Zhang
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Sergio Castro-Gonzalez
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Shuhei Nakamura
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Lars-Anders Carlson
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden,Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden,Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden,Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Dale P. Corkery
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden,Dale P. Corkery:
| | - Yao-Wen Wu
- Department of Chemistry, Umeå University, Umeå, Sweden,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden,Correspondence to Yao-Wen Wu:
| |
Collapse
|
36
|
Lei Y, Klionsky DJ. The coordination of V-ATPase and ATG16L1 is part of a common mechanism of non-canonical autophagy. Autophagy 2022; 18:2267-2269. [PMID: 35811564 PMCID: PMC9542863 DOI: 10.1080/15548627.2022.2100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/02/2022] Open
Abstract
The conjugation of Atg8-family proteins with phospholipids on the double-membrane phagophore is one of the hallmarks of macroautopahgy/autophagy. However, in the past decades, Atg8-family proteins have also been identified on single-membrane structures, including the phagosome, endosome and lysosome. While the physiological importance of the non-canonical Atg8-family protein conjugation has been demonstrated, the mechanism of this process and the underlying regulation are still not very clear. In a recent paper, Hooper et al. found that during LC3-associated phagocytosis, reactive oxygen species are required for V-ATPase assembly, which is essential for the subsequent LC3 conjugation to the phagosome. Enhanced V-ATPase assembly and the direct engagement of ATG16L1 are also observed in a wide range of non-canonical Atg8-family protein conjugation processes, defining the V-ATPase and ATG16L1 as taking part in a common mechanism.
Collapse
Affiliation(s)
- Yuchen Lei
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Control of Unconventional Secretion By The Autophagy Machinery. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
38
|
Odendall C, Sa Pessoa J, Mesquita FS. Meeting report - Cell dynamics: host-pathogen interface. J Cell Sci 2022; 135:276364. [PMID: 35979931 DOI: 10.1242/jcs.260456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two years into the most significant infectious disease event of our generation, infections have populated every conversation and in-depth understanding of host-pathogen interactions has, perhaps, never been more important. In a successful return to in-person conferences, the host-pathogen interface was the focus of the third Cell Dynamics meeting, which took place at the glorious Wotton House in Surrey, UK. The meeting organised by Michaela Gack, Maximiliano Gutierrez, Dominique Soldati-Favre and Michael Way gathered an international group of scientists who shared their recent discoveries and views on numerous aspects, including cell-autonomous defence mechanisms, pathogen interactions with host cytoskeletal or membrane dynamics, and cellular immune regulation. More than 30 years into the beginning of cellular microbiology as a field, the meeting exhibited the unique aspect of the host-pathogen interface in uncovering the fundamentals of both pathogens and their hosts.
Collapse
Affiliation(s)
- Charlotte Odendall
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, SE1 9RT London, UK
| | - Joana Sa Pessoa
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT9 7BL Belfast, UK
| | - Francisco S Mesquita
- Global Health Institute, School of Life Sciences, EPFL, CH-1015 Lausanne, Switzerland
| |
Collapse
|
39
|
Deretic V, Lazarou M. A guide to membrane atg8ylation and autophagy with reflections on immunity. J Cell Biol 2022; 221:e202203083. [PMID: 35699692 PMCID: PMC9202678 DOI: 10.1083/jcb.202203083] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 12/11/2022] Open
Abstract
The process of membrane atg8ylation, defined herein as the conjugation of the ATG8 family of ubiquitin-like proteins to membrane lipids, is beginning to be appreciated in its broader manifestations, mechanisms, and functions. Classically, membrane atg8ylation with LC3B, one of six mammalian ATG8 family proteins, has been viewed as the hallmark of canonical autophagy, entailing the formation of characteristic double membranes in the cytoplasm. However, ATG8s are now well described as being conjugated to single membranes and, most recently, proteins. Here we propose that the atg8ylation is coopted by multiple downstream processes, one of which is canonical autophagy. We elaborate on these biological outputs, which impact metabolism, quality control, and immunity, emphasizing the context of inflammation and immunological effects. In conclusion, we propose that atg8ylation is a modification akin to ubiquitylation, and that it is utilized by different systems participating in membrane stress responses and membrane remodeling activities encompassing autophagy and beyond.
Collapse
Affiliation(s)
- Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Michael Lazarou
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Hooper KM, Jacquin E, Li T, Goodwin JM, Brumell JH, Durgan J, Florey O. V-ATPase is a universal regulator of LC3-associated phagocytosis and non-canonical autophagy. J Cell Biol 2022; 221:213194. [PMID: 35511089 PMCID: PMC9082624 DOI: 10.1083/jcb.202105112] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 02/04/2022] [Accepted: 04/15/2022] [Indexed: 12/18/2022] Open
Abstract
Non-canonical autophagy is a key cellular pathway in immunity, cancer, and neurodegeneration, characterized by conjugation of ATG8 to endolysosomal single membranes (CASM). CASM is activated by engulfment (endocytosis, phagocytosis), agonists (STING, TRPML1), and infection (influenza), dependent on K490 in the ATG16L1 WD40-domain. However, factors associated with non-canonical ATG16L1 recruitment and CASM induction remain unknown. Here, using pharmacological inhibitors, we investigate a role for V-ATPase during non-canonical autophagy. We report that increased V0–V1 engagement is associated with, and sufficient for, CASM activation. Upon V0–V1 binding, V-ATPase recruits ATG16L1, via K490, during LC3-associated phagocytosis (LAP), STING- and drug-induced CASM, indicating a common mechanism. Furthermore, during LAP, key molecular players, including NADPH oxidase/ROS, converge on V-ATPase. Finally, we show that LAP is sensitive to Salmonella SopF, which disrupts the V-ATPase–ATG16L1 axis and provide evidence that CASM contributes to the Salmonella host response. Together, these data identify V-ATPase as a universal regulator of CASM and indicate that SopF evolved in part to evade non-canonical autophagy.
Collapse
Affiliation(s)
| | - Elise Jacquin
- Signalling Programme, Babraham Institute, Cambridge, UK.,Institut national de la santé et de la recherche médicale UMR-S 1193, Université Paris-Saclay, Châtenay-Malabry, France
| | - Taoyingnan Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - John H Brumell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,SickKids Inflammatory Bowel Disease Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joanne Durgan
- Signalling Programme, Babraham Institute, Cambridge, UK
| | - Oliver Florey
- Signalling Programme, Babraham Institute, Cambridge, UK
| |
Collapse
|
41
|
Münz C. Canonical and Non-Canonical Functions of the Autophagy Machinery in MHC Restricted Antigen Presentation. Front Immunol 2022; 13:868888. [PMID: 35309359 PMCID: PMC8931038 DOI: 10.3389/fimmu.2022.868888] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/15/2022] [Indexed: 01/29/2023] Open
Abstract
Macroautophagy delivers cytoplasmic constituents for lysosomal degradation. Since major histocompatibility complex (MHC) class II molecules sample peptides after lysosomal degradation for presentation to CD4+ T cells, it was originally described that these peptides can also originate from macroautophagy substrates. In recent years it has become clear that in addition to this canonical function of the macroautophagy machinery during MHC class II restricted antigen presentation at least parts of this machinery are also used to regulate phagocytosis of antigens, degradation of MHC class I molecules, and unconventional secretion of antigens in extracellular vesicles, including virus particles. This review discusses how both canonical and non-canonical functions of the macroautophagy machinery influence antigen presentation on MHC class I and II molecules to CD8+ and CD4+ T cells. A better understanding of the molecular mechanisms by which the macroautophagy machinery is distributed between its canonical and non-canonical functions should allow targeting of antigens to these different pathways to influence MHC restricted presentation during vaccination against infectious diseases and tumors.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
42
|
Timimi L, Figueras-Novoa C, Marcassa E, Florey O, Baillie JK, Beale R, Ulferts R. The V-ATPase complex regulates non-canonical Atg8-family protein lipidation through ATG16L1 recruitment. Autophagy 2022; 18:707-708. [PMID: 35258397 PMCID: PMC9037397 DOI: 10.1080/15548627.2022.2029233] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 11/02/2022] Open
Abstract
Conjugation of the Atg8 (autophagy related 8) family of ubiquitin-like proteins to phospholipids of the phagophore is a hallmark of macroautophagy/autophagy. Consequently, Atg8 family members, especially LC3B, are commonly used as a marker of autophagosomes. However, the Atg8 family of proteins are not found solely attached to double-membrane autophagosomes. In non-canonical Atg8-family protein lipidation they become conjugated to single membranes. We have shown that this process is triggered by recruitment of ATG16L1 by the vacuolar-type H+-translocating ATPase (V-ATPase) proton pump, suggesting a role for pH sensing in recruitment of Atg8-family proteins to single membranes.
Collapse
Affiliation(s)
- Lewis Timimi
- The Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
| | | | - Elena Marcassa
- The Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
| | - Oliver Florey
- Signalling Programme, The Babraham Institute, Cambridge, UK
| | | | - Rupert Beale
- The Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
- Division of Medicine, UCL, London, UK
| | - Rachel Ulferts
- The Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
43
|
Lau N, Thomas DR, Lee YW, Knodler LA, Newton HJ. Perturbation of ATG16L1 function impairs the biogenesis of Salmonella and Coxiella replication vacuoles. Mol Microbiol 2022; 117:235-251. [PMID: 34874584 PMCID: PMC8844213 DOI: 10.1111/mmi.14858] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/03/2023]
Abstract
Anti-bacterial autophagy, known as xenophagy, is a host innate immune response that targets invading pathogens for degradation. Some intracellular bacteria, such as the enteric pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium), utilize effector proteins to interfere with autophagy. One such S. Typhimurium effector, SopF, inhibits recruitment of ATG16L1 to damaged Salmonella-containing vacuoles (SCVs), thereby inhibiting the host xenophagic response. SopF is also required to maintain the integrity of the SCV during the early stages of infection. Here we show disruption of the SopF-ATG16L1 interaction leads to an increased proportion of cytosolic S. Typhimurium. Furthermore, SopF was utilized as a molecular tool to examine the requirement for ATG16L1 in the intracellular lifestyle of Coxiella burnetii, a bacterium that requires a functional autophagy pathway to replicate efficiently and form a single, spacious vacuole called the Coxiella-containing vacuole (CCV). ATG16L1 is required for CCV expansion and fusion but does not influence C. burnetii replication. In contrast, SopF did not affect CCV formation or replication, demonstrating that the contribution of ATG16L1 to CCV biogenesis is via its role in autophagy, not xenophagy. This study highlights the diverse capabilities of bacterial effector proteins to dissect the molecular details of host-pathogen interactions.
Collapse
Affiliation(s)
- Nicole Lau
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - David R Thomas
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Yi Wei Lee
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Leigh A Knodler
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia.,Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Hayley J Newton
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|