1
|
Dash BK, Urano Y, Mita Y, Ashida Y, Hirose R, Noguchi N. Unconventional secretion of PARK7 requires lysosomal delivery via chaperone-mediated autophagy and specialized SNARE complex. Proc Natl Acad Sci U S A 2025; 122:e2414790122. [PMID: 40327696 PMCID: PMC12088447 DOI: 10.1073/pnas.2414790122] [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: 07/23/2024] [Accepted: 03/09/2025] [Indexed: 05/08/2025] Open
Abstract
PARK7/DJ-1, a redox-sensitive protein implicated in neurodegeneration, cancer, and inflammation, exhibits increased secretion under stress. We previously demonstrated that, as a leaderless protein, PARK7 relies on an unconventional autophagy pathway for stress-induced secretion. The current study delves deeper into the mechanisms governing PARK7 secretion under oxidative stress triggered by the neurotoxin 6-hydroxydopamine (6-OHDA). Here, we revealed that 6-OHDA-induced autophagic flux is critical for PARK7 secretion. Downregulation of syntaxin 17 (STX17), a SNARE protein crucial for autophagosome-lysosome fusion and cargo degradation, hindered PARK7 secretion. Likewise, impairing lysosomal function with bafilomycin A1 (BafA1) or chloroquine (CQ) diminished PARK7 release, highlighting the importance of functional lysosomes, potentially in the form of secretory autolysosomes, in PARK7 release. We also found that 6-OHDA appeared to promote the unfolding of PARK7, allowing its selective recognition by the chaperone HSPA8 via KFERQ-like motifs, leading to PARK7 translocation to the lysosomal membrane through LAMP2 via chaperone-mediated autophagy (CMA). Additionally, a dedicated SNARE complex comprising Qabc-SNAREs (STX3/4, VTI1B, and STX8) and R-SNARE SEC22B mediates the fusion of PARK7-containing autolysosomes with the plasma membrane, facilitating the extracellular release of PARK7. Hence, this study uncovers a mechanism where 6-OHDA-induced autophagic flux drives the unconventional secretion of PARK7, involving CMA for PARK7 translocation to lysosomes and specialized SNARE complexes for membrane fusion events.
Collapse
Affiliation(s)
- Biplab Kumar Dash
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe610-0394, Kyoto, Japan
| | - Yasuomi Urano
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe610-0394, Kyoto, Japan
| | - Yuichiro Mita
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe610-0394, Kyoto, Japan
| | - Yuki Ashida
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe610-0394, Kyoto, Japan
| | - Ryoma Hirose
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe610-0394, Kyoto, Japan
| | - Noriko Noguchi
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe610-0394, Kyoto, Japan
| |
Collapse
|
2
|
Zhao P, Tian R, Song D, Zhu Q, Ding X, Zhang J, Cao B, Zhang M, Xu Y, Fang J, Tan J, Yi C, Xia H, Liu W, Zou W, Sun Q. Rab GTPases are evolutionarily conserved signals mediating selective autophagy. J Cell Biol 2025; 224:e202410150. [PMID: 40197538 PMCID: PMC11977514 DOI: 10.1083/jcb.202410150] [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: 11/05/2024] [Revised: 12/31/2024] [Accepted: 01/21/2025] [Indexed: 04/10/2025] Open
Abstract
Selective autophagy plays a crucial role in maintaining cellular homeostasis by specifically targeting unwanted cargo labeled with "autophagy cues" signals for autophagic degradation. In this study, we identify Rab GTPases as a class of such autophagy cues signals involved in selective autophagy. Through biochemical and imaging screens, we reveal that human Rab GTPases are common autophagy substrates. Importantly, we confirm the conservation of Rab GTPase autophagic degradation in different model organisms. Rab GTPases translocate to damaged mitochondria, lipid droplets, and invading Salmonella-containing vacuoles (SCVs) to serve as degradation signals. Furthermore, they facilitate mitophagy, lipophagy, and xenophagy, respectively, by recruiting receptors. This interplay between Rab GTPases and receptors may ensure the de novo synthesis of isolation membranes around Rab-GTPase-labeled cargo, thereby mediating selective autophagy. These processes are further influenced by upstream regulators such as LRRK2, GDIs, and RabGGTase. In conclusion, this study unveils a conserved mechanism involving Rab GTPases as autophagy cues signals and proposes a model for the spatiotemporal control of selective autophagy.
Collapse
Affiliation(s)
- Pengwei Zhao
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Rui Tian
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Dandan Song
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qi Zhu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xianming Ding
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jianqin Zhang
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Beibei Cao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengyuan Zhang
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yilu Xu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jie Fang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jieqiong Tan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Cong Yi
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongguang Xia
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Qiming Sun
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, China
| |
Collapse
|
3
|
North BJ, Ohnstad AE, Ragusa MJ, Shoemaker CJ. The LC3-interacting region of NBR1 is a protein interaction hub enabling optimal flux. J Cell Biol 2025; 224:e202407105. [PMID: 39928048 PMCID: PMC11809422 DOI: 10.1083/jcb.202407105] [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: 07/15/2024] [Revised: 12/05/2024] [Accepted: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
During autophagy, toxic cargo is encapsulated by autophagosomes and trafficked to lysosomes for degradation. NBR1, an autophagy receptor targeting ubiquitinated aggregates, serves as a model for studying the multivalent, heterotypic interactions of cargo-bound receptors. Here, we find that three critical NBR1 partners-ATG8-family proteins, FIP200, and TAX1BP1-each bind to distinct, overlapping determinants within a short linear interaction motif (SLiM). To explore whether overlapping SLiMs extend beyond NBR1, we analyzed >100 LC3-interacting regions (LIRs), revealing that FIP200 and/or TAX1BP1 binding to LIRs is a common phenomenon and suggesting LIRs as protein interaction hotspots. Phosphomimetic peptides demonstrate that phosphorylation generally enhances FIP200 and ATG8-family binding but not TAX1BP1, indicating differential regulation. In vivo, LIR-mediated interactions with TAX1BP1 promote optimal NBR1 flux by leveraging additional functionalities from TAX1BP1. These findings reveal a one-to-many binding modality in the LIR motif of NBR1, illustrating the cooperative mechanisms of autophagy receptors and the regulatory potential of multifunctional SLiMs.
Collapse
Affiliation(s)
- Brian J. North
- Department of Biochemistry and Cell Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Amelia E. Ohnstad
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | | | - Christopher J. Shoemaker
- Department of Biochemistry and Cell Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
4
|
Scavone F, Lian S, Eskelinen EL, Cohen RE, Yao T. Trafficking of K63-polyubiquitin-modified membrane proteins in a macroautophagy-independent pathway is linked to ATG9A. Mol Biol Cell 2025; 36:ar42. [PMID: 39969968 PMCID: PMC12005115 DOI: 10.1091/mbc.e24-12-0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Cytoplasmic K63-linked polyubiquitin signals have well-established roles in endocytosis and selective autophagy. However, how these signals help to direct different cargos to different intracellular trafficking routes is unclear. Here we report that, when the K63-polyubiquitin signal is blocked by intracellular expression of a high-affinity sensor (named Vx3), many proteins originating from the plasma membrane are found trapped in clusters of small vesicles that colocalize with ATG9A, a transmembrane protein that plays an essential role in autophagy. Importantly, whereas ATG9A is required for cluster formation, other core autophagy machinery as well as selective autophagy cargo receptors are not required. Although the cargos are sequestered in the vesicular clusters in an ATG9-dependent manner, additional signals are needed to induce LC3 conjugation. Upon removal of the Vx3 block, K63-polyubiquitylated cargos are rapidly delivered to lysosomes. These observations suggest that ATG9A plays an unexpected role in the trafficking of K63-polyubiquitin-modified membrane proteins.
Collapse
Affiliation(s)
- Francesco Scavone
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Sharon Lian
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Eeva-Liisa Eskelinen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, 00014, Finland
- Institute of Biomedicine, University of Turku, Turku, FI-20520, Finland
| | - Robert E. Cohen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Tingting Yao
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
5
|
Scott O, Saran E, Freeman SA. The spectrum of lysosomal stress and damage responses: from mechanosensing to inflammation. EMBO Rep 2025; 26:1425-1439. [PMID: 40016424 PMCID: PMC11933331 DOI: 10.1038/s44319-025-00405-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/01/2025] Open
Abstract
Cells and tissues turn over their aged and damaged components in order to adapt to a changing environment and maintain homeostasis. These functions rely on lysosomes, dynamic and heterogeneous organelles that play essential roles in nutrient redistribution, metabolism, signaling, gene regulation, plasma membrane repair, and immunity. Because of metabolic fluctuations and pathogenic threats, lysosomes must adapt in the short and long term to maintain functionality. In response to such challenges, lysosomes deploy a variety of mechanisms that prevent the breaching of their membrane and escape of their contents, including pathogen-associated molecules and hydrolases. While transient permeabilization of the lysosomal membrane can have acute beneficial effects, supporting inflammation and antigen cross-presentation, sustained or repeated lysosomal perforations have adverse metabolic and transcriptional consequences and can lead to cell death. This review outlines factors contributing to lysosomal stress and damage perception, as well as remedial processes aimed at addressing lysosomal disruptions. We conclude that lysosomal stress plays widespread roles in human physiology and pathology, the understanding and manipulation of which can open the door to novel therapeutic strategies.
Collapse
Affiliation(s)
- Ori Scott
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
- Division of Clinical Immunology and Allergy, Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Ekambir Saran
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Bueno-Arribas M, Cruz-Cuevas C, Monforte-Martinez B, Navas MA, Escalante R, Vincent O. The PKA Signaling Pathway Regulates the Association of the Autophagy Initiation Complex With the Lipidation Machinery. J Mol Biol 2025; 437:168954. [PMID: 39826713 DOI: 10.1016/j.jmb.2025.168954] [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: 12/02/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
A key step in autophagy is the conjugation by the E3-like Atg12-Atg5-Atg16 complex of the ubiquitin-like protein Atg8 to phosphatidylethanolamine on the autophagosomal membrane, a process known as lipidation. Previous work in yeast showed that recruitment of the E3-like complex to the preautophagosomal structure is mediated by the interaction of Atg16 with the phosphatidylinositol 3-phosphate-binding protein Atg21, and by the association of Atg12 with the scaffold protein of the Atg1 kinase complex, Atg17. Here, we conducted a reverse two-hybrid screen to identify residues in Atg17 and Atg12 critical for Atg17-Atg12 binding, and used these data to generate a docking model of Atg12-Atg5-Atg16 with the Atg17 complex. In this model, a conserved alpha-helix in the N-terminal region of Atg12 binds to the convex side of crescent-shaped Atg17 and appears to form a four-helix bundle with the three helices of Atg17, similar to that described for the binding of Atg31 to Atg17. We further showed that, in agreement with previous work, Atg17-Atg12 and Atg21-Atg16 binding act cooperatively to mediate the recruitment of the E3-like complex, although our results show that alternative mechanisms are involved in this process. Finally, we found that phosphorylation of Atg12 by PKA prevents its interaction with Atg17, thus adding a new regulatory layer in the control of autophagy by the PKA signaling pathway.
Collapse
Affiliation(s)
| | - Celia Cruz-Cuevas
- Instituto de Investigaciones Biomédicas Sols-Morreale CSIC-UAM, 28029 Madrid, Spain
| | | | - María-Angeles Navas
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Ricardo Escalante
- Instituto de Investigaciones Biomédicas Sols-Morreale CSIC-UAM, 28029 Madrid, Spain
| | - Olivier Vincent
- Instituto de Investigaciones Biomédicas Sols-Morreale CSIC-UAM, 28029 Madrid, Spain.
| |
Collapse
|
7
|
Henn D, Yang X, Li M. Lysosomal quality control Review. Autophagy 2025:1-20. [PMID: 39968899 DOI: 10.1080/15548627.2025.2469206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025] Open
Abstract
Healthy cells need functional lysosomes to degrade cargo delivered by autophagy and endocytosis. Defective lysosomes can lead to severe conditions such as lysosomal storage diseases (LSDs) and neurodegeneration. To maintain lysosome integrity and functionality, cells have evolved multiple quality control pathways corresponding to different types of stress and damage. These can be divided into five levels: regulation, reformation, repair, removal, and replacement. The different levels of lysosome quality control often work together to maintain the integrity of the lysosomal network. This review summarizes the different quality control pathways and discusses the less-studied area of lysosome membrane protein regulation and degradation, highlighting key unanswered questions in the field.Abbreviation: ALR: autophagic lysosome reformation; CASM: conjugation of ATG8 to single membranes: ER: endoplasmic reticulum; ESCRT: endosomal sorting complexes required for transport; ILF: intralumenal fragment; LSD: lysosomal storage disease; LYTL: lysosomal tubulation/sorting driven by LRRK2; PITT: phosphoinositide-initiated membrane tethering and lipid transport; PE: phosphatidylethanolamine; PLR: phagocytic lysosome reformation; PS: phosphatidylserine; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns4P: phosphatidylinositol-4-phosphate; PtdIns(4,5)P2: phosphatidylinositol-4,5-bisphosphate; V-ATPase: vacuolar-type H+-translocating ATPase.
Collapse
Affiliation(s)
- Danielle Henn
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Xi Yang
- Department of Biological Sciences, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Ming Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Yonekawa Y, Oikawa K, Bayarkhuu B, Kobayashi K, Saito N, Oikawa I, Yamada R, Chen YH, Oyanagi K, Shibasaki Y, Kobayashi S, Shiba Y. Magnetic control of membrane damage in early endosomes using internalized magnetic nanoparticles. Cell Struct Funct 2025; 50:25-39. [PMID: 39756866 DOI: 10.1247/csf.24037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Membrane stiffness is essential for cell migration, tumorigenesis, and development; however, the physical properties of intracellular membrane are poorly characterized. In this study, we internalized 20 nm magnetic nanoparticles (MNPs) into MCF7 human breast cancer cells and applied a magnetic field. We investigated whether magnetic field could induce membrane damage of the early endosomes by analyzing the colocalization of MNPs with galectin 3 (Gal3), a cytosolic protein recruited to the lumen of damaged organelles. We first tried to apply magnetic field by electromagnet, and found a direct-current (DC) magnetic field for five minutes increased the colocalization of the MNPs with Gal3, suggesting that the magnetic field damaged the endosomal membrane. We used a neodymium magnet to apply longer and stronger static magnetic fields. The static magnetic field more than 50 mT for five minutes started to damage endosomes, while 100 mT was the most effective. Longer exposure or higher magnetic field strengths did not induce further membrane damage. We confirmed that a Gal3 positive compartment was also positive for the early endosome marker, EEA1, suggesting that the external magnetic field induced membrane damage in the early endosomes. Our results indicate that a static magnetic field can control the membrane damage in early endosomes using internalized MNPs.Key words: magnetic nanoparticles, endosomes, membrane damage, organelle.
Collapse
Affiliation(s)
- Yuta Yonekawa
- Graduate Course in Biological Sciences, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Kazuki Oikawa
- Graduate Course in Biological Sciences, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Boldbaatar Bayarkhuu
- Graduate Course in Biological Sciences, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Kizuna Kobayashi
- Graduate Course in Biological Sciences, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Nana Saito
- Graduate Course in Biological Sciences, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Ibuki Oikawa
- Graduate Course in Materials Science and Engineering, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Ryohei Yamada
- Graduate Course in Materials Science and Engineering, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Yu-Han Chen
- Department of Biochemical Science and Technology, National Chiayi University
| | - Koichi Oyanagi
- Graduate Course in Materials Science and Engineering, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Yuji Shibasaki
- Graduate Course in Chemistry, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Satoru Kobayashi
- Graduate Course in Materials Science and Engineering, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| | - Yoko Shiba
- Graduate Course in Biological Sciences, Division of Science and Engineering, Graduate School of Arts and Sciences, Iwate University
| |
Collapse
|
9
|
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
|
10
|
Li X, Zhao H. Targeting secretory autophagy in solid cancers: mechanisms, immune regulation and clinical insights. Exp Hematol Oncol 2025; 14:12. [PMID: 39893499 PMCID: PMC11786567 DOI: 10.1186/s40164-025-00603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/25/2025] [Indexed: 02/04/2025] Open
Abstract
Secretory autophagy is a classical form of unconventional secretion that integrates autophagy with the secretory process, relying on highly conserved autophagy-related molecules and playing a critical role in tumor progression and treatment resistance. Traditional autophagy is responsible for degrading intracellular substances by fusing autophagosomes with lysosomes. However, secretory autophagy uses autophagy signaling to mediate the secretion of specific substances and regulate the tumor microenvironment (TME). Cytoplasmic substances are preferentially secreted rather than directed toward lysosomal degradation, involving various selective mechanisms. Moreover, substances released by secretory autophagy convey biological signals to the TME, inducing immune dysregulation and contributing to drug resistance. Therefore, elucidating the mechanisms underlying secretory autophagy is essential for improving clinical treatments. This review systematically summarizes current knowledge of secretory autophagy, from initiation to secretion, considering inter-tumor heterogeneity, explores its role across different tumor types. Furthermore, it proposes future research directions and highlights unresolved clinical challenges.
Collapse
Affiliation(s)
- Xinyu Li
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China
| | - Haiying Zhao
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang City, 110032, Liaoning Province, China.
| |
Collapse
|
11
|
Domingues N, Catarino S, Cristóvão B, Rodrigues L, Carvalho FA, Sarmento MJ, Zuzarte M, Almeida J, Ribeiro-Rodrigues T, Correia-Rodrigues Â, Fernandes F, Rodrigues-Santos P, Aasen T, Santos NC, Korolchuk VI, Gonçalves T, Milosevic I, Raimundo N, Girão H. Connexin43 promotes exocytosis of damaged lysosomes through actin remodelling. EMBO J 2024; 43:3627-3649. [PMID: 39044100 PMCID: PMC11377567 DOI: 10.1038/s44318-024-00177-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
A robust and efficient cellular response to lysosomal membrane damage prevents leakage from the lysosome lumen into the cytoplasm. This response is understood to happen through either lysosomal membrane repair or lysophagy. Here we report exocytosis as a third response mechanism to lysosomal damage, which is further potentiated when membrane repair or lysosomal degradation mechanisms are impaired. We show that Connexin43 (Cx43), a protein canonically associated with gap junctions, is recruited from the plasma membrane to damaged lysosomes, promoting their secretion and accelerating cell recovery. The effects of Cx43 on lysosome exocytosis are mediated by a reorganization of the actin cytoskeleton that increases plasma membrane fluidity and decreases cell stiffness. Furthermore, we demonstrate that Cx43 interacts with the actin nucleator Arp2, the activity of which was shown to be necessary for Cx43-mediated actin rearrangement and lysosomal exocytosis following damage. These results define a novel mechanism of lysosomal quality control whereby Cx43-mediated actin remodelling potentiates the secretion of damaged lysosomes.
Collapse
Affiliation(s)
- Neuza Domingues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Steve Catarino
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Beatriz Cristóvão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria João Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Jani Almeida
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Teresa Ribeiro-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Ânia Correia-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Fábio Fernandes
- Institute for Bioengineering and Biosciences (IBB) and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo Rodrigues-Santos
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Trond Aasen
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Teresa Gonçalves
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- University of Oxford, Centre for Human Genetics, Nuffield Department of Medicine, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Henrique Girão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal.
| |
Collapse
|
12
|
Gubas A, Attridge E, Jefferies HB, Nishimura T, Razi M, Kunzelmann S, Gilad Y, Mercer TJ, Wilson MM, Kimchi A, Tooze SA. WIPI2b recruitment to phagophores and ATG16L1 binding are regulated by ULK1 phosphorylation. EMBO Rep 2024; 25:3789-3811. [PMID: 39152217 PMCID: PMC11387628 DOI: 10.1038/s44319-024-00215-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 08/19/2024] Open
Abstract
One of the key events in autophagy is the formation of a double-membrane phagophore, and many regulatory mechanisms underpinning this remain under investigation. WIPI2b is among the first proteins to be recruited to the phagophore and is essential for stimulating autophagy flux by recruiting the ATG12-ATG5-ATG16L1 complex, driving LC3 and GABARAP lipidation. Here, we set out to investigate how WIPI2b function is regulated by phosphorylation. We studied two phosphorylation sites on WIPI2b, S68 and S284. Phosphorylation at these sites plays distinct roles, regulating WIPI2b's association with ATG16L1 and the phagophore, respectively. We confirm WIPI2b is a novel ULK1 substrate, validated by the detection of endogenous phosphorylation at S284. Notably, S284 is situated within an 18-amino acid stretch, which, when in contact with liposomes, forms an amphipathic helix. Phosphorylation at S284 disrupts the formation of the amphipathic helix, hindering the association of WIPI2b with membranes and autophagosome formation. Understanding these intricacies in the regulatory mechanisms governing WIPI2b's association with its interacting partners and membranes, holds the potential to shed light on these complex processes, integral to phagophore biogenesis.
Collapse
Affiliation(s)
- Andrea Gubas
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Muscular Dystrophy UK, London, SE1 8QD, UK
| | - Eleanor Attridge
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Harold Bj Jefferies
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Taki Nishimura
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| | - Minoo Razi
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Simone Kunzelmann
- Structural Biology Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Yuval Gilad
- The Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Adi Kimchi
- The Weizmann Institute of Science, Rehovot, Israel
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
13
|
Yanagawa K, Kuma A, Hamasaki M, Kita S, Yamamuro T, Nishino K, Nakamura S, Omori H, Kaminishi T, Oikawa S, Kato Y, Edahiro R, Kawagoe R, Taniguchi T, Tanaka Y, Shima T, Tabata K, Iwatani M, Bekku N, Hanayama R, Okada Y, Akimoto T, Kosako H, Takahashi A, Shimomura I, Sakata Y, Yoshimori T. The Rubicon-WIPI axis regulates exosome biogenesis during ageing. Nat Cell Biol 2024; 26:1558-1570. [PMID: 39174742 DOI: 10.1038/s41556-024-01481-0] [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: 08/28/2023] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
Cells release intraluminal vesicles in multivesicular bodies as exosomes to communicate with other cells. Although recent studies suggest an intimate link between exosome biogenesis and autophagy, the detailed mechanism is not fully understood. Here we employed comprehensive RNA interference screening for autophagy-related factors and discovered that Rubicon, a negative regulator of autophagy, is essential for exosome release. Rubicon recruits WIPI2d to endosomes to promote exosome biogenesis. Interactome analysis of WIPI2d identified the ESCRT components that are required for intraluminal vesicle formation. Notably, we found that Rubicon is required for an age-dependent increase of exosome release in mice. In addition, small RNA sequencing of serum exosomes revealed that Rubicon determines the fate of exosomal microRNAs associated with cellular senescence and longevity pathways. Taken together, our current results suggest that the Rubicon-WIPI axis functions as a key regulator of exosome biogenesis and is responsible for age-dependent changes in exosome quantity and quality.
Collapse
Affiliation(s)
- Kyosuke Yanagawa
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akiko Kuma
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
- Health Promotion System Science, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Maho Hamasaki
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
- Tokyo New Drug Research Laboratories, Pharmaceutical Business Unit, Kowa Company, Higashimurayama, Japan
| | - Tadashi Yamamuro
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Kohei Nishino
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Shuhei Nakamura
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Suita, Japan
- Department of Biochemistry, Nara Medical University, Kashihara, Japan
| | - Hiroko Omori
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tatsuya Kaminishi
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Satoshi Oikawa
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Waseda Institute for Sport Sciences, Waseda University, Tokorozawa, Japan
| | - Yoshio Kato
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryosuke Kawagoe
- i2i-Labo, Yokohama Research Center, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama, Japan
| | - Takako Taniguchi
- i2i-Labo, Yokohama Research Center, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama, Japan
| | - Yoko Tanaka
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takayuki Shima
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Biochemistry, Nara Medical University, Kashihara, Japan
| | - Keisuke Tabata
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Miki Iwatani
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Nao Bekku
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Rikinari Hanayama
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- WPI Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita, Japan
| | - Takayuki Akimoto
- Laboratory of Muscle Biology, Faculty of Sport Sciences, Waseda University, Tokorozawa, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Akiko Takahashi
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan.
- Health Promotion System Science, Graduate School of Medicine, Osaka University, Suita, Japan.
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan.
| |
Collapse
|
14
|
Wu X, Yang Y, Ru Y, Hao R, Zhao D, Ren R, Lu B, Li Y, Sun S, Zheng H, Wang W. Knockout of the WD40 domain of ATG16L1 enhances foot and mouth disease virus replication. BMC Genomics 2024; 25:796. [PMID: 39179961 PMCID: PMC11342673 DOI: 10.1186/s12864-024-10703-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024] Open
Abstract
The WD40 domain is one of the most abundant domains and is among the top interacting domains in eukaryotic genomes. The WD40 domain of ATG16L1 is essential for LC3 recruitment to endolysosomal membranes during non-canonical autophagy, but dispensable for canonical autophagy. Canonical autophagy was utilized by FMDV, while the relationship between FMDV and non-canonical autophagy is still elusive. In the present study, WD40 knockout (KO) PK15 cells were successfully generated via CRISPR/cas9 technology as a tool for studying the effect of non-canonical autophagy on FMDV replication. The results of growth curve analysis, morphological observation and karyotype analysis showed that the WD40 knockout cell line was stable in terms of growth and morphological characteristics. After infection with FMDV, the expression of viral protein, viral titers, and the number of copies of viral RNA in the WD40-KO cells were significantly greater than those in the wild-type PK15 cells. Moreover, RNA‒seq technology was used to sequence WD40-KO cells and wild-type cells infected or uninfected with FMDV. Differentially expressed factors such as Mx1, RSAD2, IFIT1, IRF9, IFITM3, GBP1, CXCL8, CCL5, TNFRSF17 were significantly enriched in the autophagy, NOD-like receptor signaling pathway, RIG-I-like receptor signaling pathway, Toll-like receptor signaling pathway, cytokine-cytokine receptor interaction and TNF signaling pathway, etc. The expression levels of differentially expressed genes were detected via qRT‒PCR, which was consistent with the RNA‒seq data. Here, we experimentally demonstrate for the first time that knockout of the WD40 domain of ATG16L1 enhances FMDV replication by downregulation innate immune factors. In addition, this result also indicates non-canonical autophagy inhibits FMDV replication. In total, our results play an essential role in regulating the replication level of FMDV and providing new insights into virus-host interactions and potential antiviral strategies.
Collapse
Affiliation(s)
- Xiuping Wu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Yang Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Yi Ru
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Rongzeng Hao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Dongmei Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Ruifang Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Bingzhou Lu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Yajun Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Shengzhen Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Wenhui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
15
|
Eickhorst C, Babic R, Rush-Kittle J, Lucya L, Imam FL, Sánchez-Martín P, Hollenstein DM, Michaelis J, Münch C, Meisinger C, Slade D, Gámez-Díaz L, Kraft C. FIP200 Phosphorylation Regulates Late Steps in Mitophagy. J Mol Biol 2024; 436:168631. [PMID: 38821350 DOI: 10.1016/j.jmb.2024.168631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Mitophagy is a specific type of autophagy responsible for the selective elimination of dysfunctional or superfluous mitochondria, ensuring the maintenance of mitochondrial quality control. The initiation of mitophagy is coordinated by the ULK1 kinase complex, which engages mitophagy receptors via its FIP200 subunit. Whether FIP200 performs additional functions in the subsequent later phases of mitophagy beyond this initial step and how its regulation occurs, remains unclear. Our findings reveal that multiple phosphorylation events on FIP200 differentially control the early and late stages of mitophagy. Furthermore, these phosphorylation events influence FIP200's interaction with ATG16L1. In summary, our results highlight the necessity for precise and dynamic regulation of FIP200, underscoring its importance in the progression of mitophagy.
Collapse
Affiliation(s)
- Christopher Eickhorst
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Riccardo Babic
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Jorrell Rush-Kittle
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Leon Lucya
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Fatimah Lami Imam
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Pablo Sánchez-Martín
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - David M Hollenstein
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Department for Biochemistry and Cell Biology, University of Vienna, Center for Molecular Biology, Vienna Biocenter Campus (VBC), Dr. Bohr-Gasse 9, 1030 Vienna, Austria; Mass Spectrometry Facility, Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr. Bohr-Gasse 7, 1030 Vienna, Austria
| | - Jonas Michaelis
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Christian Münch
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Chris Meisinger
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Dea Slade
- Department of Radiation Oncology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - Laura Gámez-Díaz
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| | - Claudine Kraft
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
16
|
Prashar A, Bussi C, Fearns A, Capurro MI, Gao X, Sesaki H, Gutierrez MG, Jones NL. Lysosomes drive the piecemeal removal of mitochondrial inner membrane. Nature 2024; 632:1110-1117. [PMID: 39169179 PMCID: PMC7616637 DOI: 10.1038/s41586-024-07835-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Mitochondrial membranes define distinct structural and functional compartments. Cristae of the inner mitochondrial membrane (IMM) function as independent bioenergetic units that undergo rapid and transient remodelling, but the significance of this compartmentalized organization is unknown1. Using super-resolution microscopy, here we show that cytosolic IMM vesicles, devoid of outer mitochondrial membrane or mitochondrial matrix, are formed during resting state. These vesicles derived from the IMM (VDIMs) are formed by IMM herniation through pores formed by voltage-dependent anion channel 1 in the outer mitochondrial membrane. Live-cell imaging showed that lysosomes in proximity to mitochondria engulfed the herniating IMM and, aided by the endosomal sorting complex required for transport machinery, led to the formation of VDIMs in a microautophagy-like process, sparing the remainder of the organelle. VDIM formation was enhanced in mitochondria undergoing oxidative stress, suggesting their potential role in maintenance of mitochondrial function. Furthermore, the formation of VDIMs required calcium release by the reactive oxygen species-activated, lysosomal calcium channel, transient receptor potential mucolipin 1, showing an interorganelle communication pathway for maintenance of mitochondrial homeostasis. Thus, IMM compartmentalization could allow for the selective removal of damaged IMM sections via VDIMs, which should protect mitochondria from localized injury. Our findings show a new pathway of intramitochondrial quality control.
Collapse
Affiliation(s)
- Akriti Prashar
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- NHLBI, NIH, Bethesda, MD, USA
| | - Claudio Bussi
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
- School of Biological Sciences, Nanyang Technical University, Singapore, Singapore
| | - Antony Fearns
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
| | - Mariana I Capurro
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaodong Gao
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
| | - Nicola L Jones
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Departments of Paediatrics and Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
17
|
Chen P, Cao XW, Dong JW, Zhao J, Wang FJ. Saponin and Ribosome-Inactivating Protein Synergistically Trigger Lysosome-Dependent Apoptosis by Inhibiting Lysophagy: Potential to Become a New Antitumor Strategy. Mol Pharm 2024; 21:2993-3005. [PMID: 38722865 DOI: 10.1021/acs.molpharmaceut.4c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The susceptibility of lysosomal membranes in tumor cells to cationic amphiphilic drugs (CADs) enables CADs to induce lysosomal membrane permeabilization (LMP) and trigger lysosome-dependent cell death (LDCD), suggesting a potential antitumor therapeutic approach. However, the existence of intrinsic lysosomal damage response mechanisms limits the display of the pharmacological activity of CADs. In this study, we report that low concentrations of QS-21, a saponin with cationic amphiphilicity extracted from Quillaja Saponaria tree, can induce LMP but has nontoxicity to tumor cells. QS-21 and MAP30, a type I ribosome-inactivating protein, synergistically induce apoptosis in tumor cells at low concentrations of both. Mechanistically, QS-21-induced LMP helps MAP30 escape from endosomes or lysosomes and subsequently enter the endoplasmic reticulum, where MAP30 downregulates the expression of autophagy-associated LC3 proteins, thereby inhibiting lysophagy. The inhibition of lysophagy results in the impaired clearance of damaged lysosomes, leading to the leakage of massive lysosomal contents such as cathepsins into the cytoplasm, ultimately triggering LDCD. In summary, our study showed that coadministration of QS-21 and MAP30 amplified the lysosomal disruption and can be a new synergistic LDCD-based antitumor therapy.
Collapse
Affiliation(s)
- Piao Chen
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Xue-Wei Cao
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
| | - Jing-Wen Dong
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
| | - Jian Zhao
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Fu-Jun Wang
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| |
Collapse
|
18
|
North BJ, Ohnstad AE, Ragusa MJ, Shoemaker CJ. The LC3-interacting region of NBR1 is a protein interaction hub enabling optimal flux. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593318. [PMID: 38766171 PMCID: PMC11100792 DOI: 10.1101/2024.05.09.593318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
During autophagy, potentially toxic cargo is enveloped by a newly formed autophagosome and trafficked to the lysosome for degradation. Ubiquitinated protein aggregates, a key target for autophagy, are identified by multiple autophagy receptors. NBR1 is an archetypal autophagy receptor and an excellent model for deciphering the role of the multivalent, heterotypic interactions made by cargo-bound receptors. Using NBR1 as a model, we find that three critical binding partners - ATG8-family proteins, FIP200, and TAX1BP1 - each bind to a short linear interaction motif (SLiM) within NBR1. Mutational peptide arrays indicate that these binding events are mediated by distinct overlapping determinants, rather than a single, convergent, SLiM. AlphaFold modeling underlines the need for conformational flexibility within the NBR1 SLiM, as distinct conformations mediate each binding event. To test the extent to which overlapping SLiMs exist beyond NBR1, we performed peptide binding arrays on >100 established LC3-interacting regions (LIRs), revealing that FIP200 and/or TAX1BP1 binding to LIRs is a common phenomenon and suggesting LIRs as protein interaction hotspots. Comparative analysis of phosphomimetic peptides highlights that while FIP200 and Atg8-family binding are generally augmented by phosphorylation, TAX1BP1 binding is nonresponsive, suggesting differential regulation of these binding events. In vivo studies confirm that LIR-mediated interactions with TAX1BP1 enhance NBR1 activity, increasing autophagosomal delivery by leveraging an additional LIR from TAX1BP1. In sum, these results reveal a one-to-many binding modality in NBR1, providing key insights into the cooperative mechanisms among autophagy receptors. Furthermore, these findings underscore the pervasive role of multifunctional SLiMs in autophagy, offering substantial avenues for further exploration into their regulatory functions.
Collapse
Affiliation(s)
- Brian J North
- Department of Biochemistry and Cell Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Amelia E Ohnstad
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | | | - Christopher J Shoemaker
- Department of Biochemistry and Cell Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
19
|
Dashti Z, Yousefi Z, Kiani P, Taghizadeh M, Maleki MH, Borji M, Vakili O, Shafiee SM. Autophagy and the unfolded protein response shape the non-alcoholic fatty liver landscape: decoding the labyrinth. Metabolism 2024; 154:155811. [PMID: 38309690 DOI: 10.1016/j.metabol.2024.155811] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/23/2024] [Accepted: 01/28/2024] [Indexed: 02/05/2024]
Abstract
The incidence of nonalcoholic fatty liver disease (NAFLD) is on the rise, mirroring a global surge in diabetes and metabolic syndrome, as its major leading causes. NAFLD represents a spectrum of liver disorders, ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH), which can potentially progress to cirrhosis and hepatocellular carcinoma (HCC). Mechanistically, we know the unfolded protein response (UPR) as a protective cellular mechanism, being triggered under circumstances of endoplasmic reticulum (ER) stress. The hepatic UPR is turned on in a broad spectrum of liver diseases, including NAFLD. Recent data also defines molecular mechanisms that may underlie the existing correlation between UPR activation and NAFLD. More interestingly, subsequent studies have demonstrated an additional mechanism, i.e. autophagy, to be involved in hepatic steatosis, and thus NAFLD pathogenesis, principally by regulating the insulin sensitivity, hepatocellular injury, innate immunity, fibrosis, and carcinogenesis. All these findings suggest possible mechanistic roles for autophagy in the progression of NAFLD and its complications. Both UPR and autophagy are dynamic and interconnected fluxes that act as protective responses to minimize the harmful effects of hepatic lipid accumulation, as well as the ER stress during NAFLD. The functions of UPR and autophagy in the liver, together with findings of decreased hepatic autophagy in correlation with conditions that predispose to NAFLD, such as obesity and aging, suggest that autophagy and UPR, alone or combined, may be novel therapeutic targets against the disease. In this review, we discuss the current evidence on the interplay between autophagy and the UPR in connection to the NAFLD pathogenesis.
Collapse
Affiliation(s)
- Zahra Dashti
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zeynab Yousefi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Motahareh Taghizadeh
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hasan Maleki
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Borji
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
20
|
Gahlot P, Kravic B, Rota G, van den Boom J, Levantovsky S, Schulze N, Maspero E, Polo S, Behrends C, Meyer H. Lysosomal damage sensing and lysophagy initiation by SPG20-ITCH. Mol Cell 2024; 84:1556-1569.e10. [PMID: 38503285 DOI: 10.1016/j.molcel.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/30/2024] [Accepted: 02/27/2024] [Indexed: 03/21/2024]
Abstract
Cells respond to lysosomal membrane permeabilization by membrane repair or selective macroautophagy of damaged lysosomes, termed lysophagy, but it is not fully understood how this decision is made. Here, we uncover a pathway in human cells that detects lipid bilayer perturbations in the limiting membrane of compromised lysosomes, which fail to be repaired, and then initiates ubiquitin-triggered lysophagy. We find that SPG20 binds the repair factor IST1 on damaged lysosomes and, importantly, integrates that with the detection of damage-associated lipid-packing defects of the lysosomal membrane. Detection occurs via sensory amphipathic helices in SPG20 before rupture of the membrane. If lipid-packing defects are extensive, such as during lipid peroxidation, SPG20 recruits and activates ITCH, which marks the damaged lysosome with lysine-63-linked ubiquitin chains to initiate lysophagy and thus triages the lysosome for destruction. With SPG20 being linked to neurodegeneration, these findings highlight the relevance of a coordinated lysosomal damage response for cellular homeostasis.
Collapse
Affiliation(s)
- Pinki Gahlot
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Bojana Kravic
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Giulia Rota
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Johannes van den Boom
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Sophie Levantovsky
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Nina Schulze
- Imaging Center Campus Essen, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Elena Maspero
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Simona Polo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Christian Behrends
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Hemmo Meyer
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
21
|
Klickstein JA, Johnson MA, Antonoudiou P, Maguire J, Paulo JA, Gygi SP, Weihl C, Raman M. ALS-related p97 R155H mutation disrupts lysophagy in iPSC-derived motor neurons. Stem Cell Reports 2024; 19:366-382. [PMID: 38335961 PMCID: PMC10937112 DOI: 10.1016/j.stemcr.2024.01.002] [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: 07/11/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/12/2024] Open
Abstract
Mutations in the AAA+ ATPase p97 cause multisystem proteinopathy 1, which includes amyotrophic lateral sclerosis; however, the pathogenic mechanisms that contribute to motor neuron loss remain obscure. Here, we use two induced pluripotent stem cell models differentiated into spinal motor neurons to investigate how p97 mutations perturb the motor neuron proteome. Using quantitative proteomics, we find that motor neurons harboring the p97 R155H mutation have deficits in the selective autophagy of lysosomes (lysophagy). p97 R155H motor neurons are unable to clear damaged lysosomes and have reduced viability. Lysosomes in mutant motor neurons have increased pH compared with wild-type cells. The clearance of damaged lysosomes involves UBXD1-p97 interaction, which is disrupted in mutant motor neurons. Finally, inhibition of the ATPase activity of p97 using the inhibitor CB-5083 rescues lysophagy defects in mutant motor neurons. These results add to the evidence that endo-lysosomal dysfunction is a key aspect of disease pathogenesis in p97-related disorders.
Collapse
Affiliation(s)
- Jacob A Klickstein
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA
| | - Michelle A Johnson
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA
| | | | - Jamie Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Chris Weihl
- Department of Neurology, Washington University at St. Louis, St. Louis, MO
| | - Malavika Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA.
| |
Collapse
|
22
|
Gheitasi H, Sabbaghian M, Fadaee M, Mohammadzadeh N, Shekarchi AA, Poortahmasebi V. The relationship between autophagy and respiratory viruses. Arch Microbiol 2024; 206:136. [PMID: 38436746 DOI: 10.1007/s00203-024-03838-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 03/05/2024]
Abstract
Respiratory viruses have caused severe global health problems and posed essential challenges to the medical community. In recent years, the role of autophagy as a critical process in cells in viral respiratory diseases has been noticed. One of the vital catabolic biological processes in the body is autophagy. Autophagy contributes to energy recovery by targeting and selectively directing foreign microorganisms, organelles, and senescent intracellular proteins to the lysosome for degradation and phagocytosis. Activation or suppression of autophagy is often initiated when foreign pathogenic organisms such as viruses infect cells. Because of its antiviral properties, several viruses may escape or resist this process by encoding viral proteins. Viruses can also use autophagy to enhance their replication or prolong the persistence of latent infections. Here, we provide an overview of autophagy and respiratory viruses such as coronavirus, rhinovirus, parainfluenza, influenza, adenovirus, and respiratory syncytial virus, and examine the interactions between them and the role of autophagy in the virus-host interaction process and the resulting virus replication strategy.
Collapse
Affiliation(s)
- Hamidreza Gheitasi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sabbaghian
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader Mohammadzadeh
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Settembre C, Perera RM. Lysosomes as coordinators of cellular catabolism, metabolic signalling and organ physiology. Nat Rev Mol Cell Biol 2024; 25:223-245. [PMID: 38001393 DOI: 10.1038/s41580-023-00676-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/26/2023]
Abstract
Every cell must satisfy basic requirements for nutrient sensing, utilization and recycling through macromolecular breakdown to coordinate programmes for growth, repair and stress adaptation. The lysosome orchestrates these key functions through the synchronised interplay between hydrolytic enzymes, nutrient transporters and signalling factors, which together enable metabolic coordination with other organelles and regulation of specific gene expression programmes. In this Review, we discuss recent findings on lysosome-dependent signalling pathways, focusing on how the lysosome senses nutrient availability through its physical and functional association with mechanistic target of rapamycin complex 1 (mTORC1) and how, in response, the microphthalmia/transcription factor E (MiT/TFE) transcription factors exert feedback regulation on lysosome biogenesis. We also highlight the emerging interactions of lysosomes with other organelles, which contribute to cellular homeostasis. Lastly, we discuss how lysosome dysfunction contributes to diverse disease pathologies and how inherited mutations that compromise lysosomal hydrolysis, transport or signalling components lead to multi-organ disorders with severe metabolic and neurological impact. A deeper comprehension of lysosomal composition and function, at both the cellular and organismal level, may uncover fundamental insights into human physiology and disease.
Collapse
Affiliation(s)
- Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| | - Rushika M Perera
- Department of Anatomy, University of California at San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Ebstrup ML, Sønder SL, Fogde DL, Heitmann ASB, Dietrich TN, Dias C, Jäättelä M, Maeda K, Nylandsted J. Annexin A7 mediates lysosome repair independently of ESCRT-III. Front Cell Dev Biol 2024; 11:1211498. [PMID: 38348092 PMCID: PMC10860759 DOI: 10.3389/fcell.2023.1211498] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/21/2023] [Indexed: 02/15/2024] Open
Abstract
Lysosomes are crucial organelles essential for various cellular processes, and any damage to them can severely compromise cell viability. This study uncovers a previously unrecognized function of the calcium- and phospholipid-binding protein Annexin A7 in lysosome repair, which operates independently of the Endosomal Sorting Complex Required for Transport (ESCRT) machinery. Our research reveals that Annexin A7 plays a role in repairing damaged lysosomes, different from its role in repairing the plasma membrane, where it facilitates repair through the recruitment of ESCRT-III components. Notably, our findings strongly suggest that Annexin A7, like the ESCRT machinery, is dispensable for membrane contact site formation within the newly discovered phosphoinositide-initiated membrane tethering and lipid transport (PITT) pathway. Instead, we speculate that Annexin A7 is recruited to damaged lysosomes and promotes repair through its membrane curvature and cross-linking capabilities. Our findings provide new insights into the diverse mechanisms underlying lysosomal membrane repair and highlight the multifunctional role of Annexin A7 in membrane repair.
Collapse
Affiliation(s)
| | | | | | | | | | - Catarina Dias
- Membrane Integrity, Danish Cancer Institute, Copenhagen, Denmark
| | - Marja Jäättelä
- Cell Death and Metabolism, Danish Cancer Institute, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kenji Maeda
- Cell Death and Metabolism, Danish Cancer Institute, Copenhagen, Denmark
| | - Jesper Nylandsted
- Membrane Integrity, Danish Cancer Institute, Copenhagen, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
25
|
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: 33] [Impact Index Per Article: 16.5] [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
|
26
|
Zang L, Yang X, Chen Y, Huang F, Yuan Y, Chen X, Zuo Y, Miao Y, Gu J, Guo H, Xia W, Peng Y, Tang M, Huang Z, Wang Y, Ma J, Jiang J, Zhou W, Zheng H, Shi W. Ubiquitin E3 ligase SPOP is a host negative regulator of enterovirus 71-encoded 2A protease. J Virol 2023; 97:e0078623. [PMID: 37796126 PMCID: PMC10617436 DOI: 10.1128/jvi.00786-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/25/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE EV71 poses a significant health threat to children aged 5 and below. The process of EV71 infection and replication is predominantly influenced by ubiquitination modifications. Our previous findings indicate that EV71 prompts the activation of host deubiquitinating enzymes, thereby impeding the host interferon signaling pathway as a means of evading the immune response. Nevertheless, the precise mechanisms by which the host employs ubiquitination modifications to hinder EV71 infection remain unclear. The present study demonstrated that the nonstructural protein 2Apro, which is encoded by EV71, exhibits ubiquitination and degradation mediated by the host E3 ubiquitin ligase SPOP. In addition, it is the first report, to our knowledge, that SPOP is involved in the host antiviral response.
Collapse
Affiliation(s)
- Lichao Zang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Xinyu Yang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yan Chen
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Fan Huang
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yukang Yuan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Xiangjie Chen
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yibo Zuo
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Ying Miao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Jin Gu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Hui Guo
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Wenxin Xia
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yang Peng
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Mengyuan Tang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ziwei Huang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yangyang Wang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jinhong Ma
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Wei Zhou
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Hui Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Weifeng Shi
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
27
|
Corkery DP, Castro‐Gonzalez S, Knyazeva A, Herzog LK, Wu Y. An ATG12-ATG5-TECPR1 E3-like complex regulates unconventional LC3 lipidation at damaged lysosomes. EMBO Rep 2023; 24:e56841. [PMID: 37381828 PMCID: PMC10481663 DOI: 10.15252/embr.202356841] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/30/2023] Open
Abstract
Lysosomal membrane damage represents a threat to cell viability. As such, cells have evolved sophisticated mechanisms to maintain lysosomal integrity. Small membrane lesions are detected and repaired by the endosomal sorting complex required for transport (ESCRT) machinery while more extensively damaged lysosomes are cleared by a galectin-dependent selective macroautophagic pathway (lysophagy). In this study, we identify a novel role for the autophagosome-lysosome tethering factor, TECPR1, in lysosomal membrane repair. Lysosomal damage promotes TECPR1 recruitment to damaged membranes via its N-terminal dysferlin domain. This recruitment occurs upstream of galectin and precedes the induction of lysophagy. At the damaged membrane, TECPR1 forms an alternative E3-like conjugation complex with the ATG12-ATG5 conjugate to regulate ATG16L1-independent unconventional LC3 lipidation. Abolishment of LC3 lipidation via ATG16L1/TECPR1 double knockout impairs lysosomal recovery following damage.
Collapse
Affiliation(s)
- Dale P Corkery
- Department of ChemistryUmeå UniversityUmeåSweden
- Umeå Centre for Microbial ResearchUmeå UniversityUmeåSweden
| | - Sergio Castro‐Gonzalez
- Department of ChemistryUmeå UniversityUmeåSweden
- Umeå Centre for Microbial ResearchUmeå UniversityUmeåSweden
| | - Anastasia Knyazeva
- Department of ChemistryUmeå UniversityUmeåSweden
- Umeå Centre for Microbial ResearchUmeå UniversityUmeåSweden
| | - Laura K Herzog
- Department of ChemistryUmeå UniversityUmeåSweden
- Umeå Centre for Microbial ResearchUmeå UniversityUmeåSweden
| | - Yao‐Wen Wu
- Department of ChemistryUmeå UniversityUmeåSweden
- Umeå Centre for Microbial ResearchUmeå UniversityUmeåSweden
| |
Collapse
|
28
|
Yang H, Tan JX. Lysosomal quality control: molecular mechanisms and therapeutic implications. Trends Cell Biol 2023; 33:749-764. [PMID: 36717330 PMCID: PMC10374877 DOI: 10.1016/j.tcb.2023.01.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/29/2023]
Abstract
Lysosomes are essential catabolic organelles with an acidic lumen and dozens of hydrolytic enzymes. The detrimental consequences of lysosomal leakage have been well known since lysosomes were discovered during the 1950s. However, detailed knowledge of lysosomal quality control mechanisms has only emerged relatively recently. It is now clear that lysosomal leakage triggers multiple lysosomal quality control pathways that replace, remove, or directly repair damaged lysosomes. Here, we review how lysosomal damage is sensed and resolved in mammalian cells, with a focus on the molecular mechanisms underlying different lysosomal quality control pathways. We also discuss the clinical implications and therapeutic potential of these pathways.
Collapse
Affiliation(s)
- Haoxiang Yang
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA
| | - Jay Xiaojun Tan
- Aging Institute, University of Pittsburgh School of Medicine/University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
29
|
Bai Y, Yu G, Zhou HM, Amarasinghe O, Zhou Y, Zhu P, Li Q, Zhang L, Nguele Meke F, Miao Y, Chapman E, Tao WA, Zhang ZY. PTP4A2 promotes lysophagy by dephosphorylation of VCP/p97 at Tyr805. Autophagy 2023; 19:1562-1581. [PMID: 36300783 PMCID: PMC10240998 DOI: 10.1080/15548627.2022.2140558] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 11/02/2022] Open
Abstract
Overexpression of PTP4A phosphatases are associated with advanced cancers, but their biological functions are far from fully understood due to limited knowledge about their physiological substrates. VCP is implicated in lysophagy via collaboration with specific cofactors in the ELDR complex. However, how the ELDR complex assembly is regulated has not been determined. Moreover, the functional significance of the penultimate and conserved Tyr805 phosphorylation in VCP has not been established. Here, we use an unbiased substrate trapping and mass spectrometry approach and identify VCP/p97 as a bona fide substrate of PTP4A2. Biochemical studies show that PTP4A2 dephosphorylates VCP at Tyr805, enabling the association of VCP with its C-terminal cofactors UBXN6/UBXD1 and PLAA, which are components of the ELDR complex responsible for lysophagy, the autophagic clearance of damaged lysosomes. Functionally, PTP4A2 is required for cellular homeostasis by promoting lysophagy through facilitating ELDR-mediated K48-linked ubiquitin conjugate removal and autophagosome formation on the damaged lysosomes. Deletion of Ptp4a2 in vivo compromises the recovery of glycerol-injection induced acute kidney injury due to impaired lysophagy and sustained lysosomal damage. Taken together, our data establish PTP4A2 as a critical regulator of VCP and uncover an important role for PTP4A2 in maintaining lysosomal homeostasis through dephosphorylation of VCP at Tyr805. Our study suggests that PTP4A2 targeting could be a potential therapeutic approach to treat cancers and other degenerative diseases by modulating lysosomal homeostasis and macroautophagy/autophagy.Abbreviations: AAA+: ATPases associated with diverse cellular activities; AKI: acute kidney injury; CBB: Coomassie Brilliant Blue; CRISPR: clustered regularly interspaced short palindromic repeats; ELDR: endo-lysosomal damage response; GFP: green fluorescent protein; GST: glutathione S-transferase; IHC: immunohistochemistry; IP: immunoprecipitation; LAMP1: lysosomal-associated membrane protein 1; LC-MS: liquid chromatography-mass spectrometry; LGALS3/Gal3: galectin 3; LLOMe: L-leucyl-L-leucine methyl ester; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; PLAA: phospholipase A2, activating protein; PTP4A2: protein tyrosine phosphatase 4a2; PUB: NGLY1/PNGase/UBA- or UBX-containing protein; PUL: PLAP, Ufd3, and Lub1; TFEB: transcription factor EB; UBXN6/UBXD1: UBX domain protein 6; UPS: ubiquitin-proteasome system; VCP/p97: valosin containing protein; VCPIP1: valosin containing protein interacting protein 1; YOD1: YOD1 deubiquitinase.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, USA
| | - Guimei Yu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, USA
| | - Hong-Ming Zhou
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Yuan Zhou
- Department of Biochemistry, Purdue University, West Lafayette, USA
| | - Peipei Zhu
- Department of Chemistry, Purdue University, West Lafayette, USA
| | - Qinglin Li
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, USA
| | - Lujuan Zhang
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, IN, USA
| | - Frederick Nguele Meke
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, USA
| | - Yiming Miao
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, A, USA
| | - W. Andy Tao
- Department of Chemistry, Purdue University, West Lafayette, USA
- Department of Biochemistry, Purdue University, West Lafayette, USA
- Center for Cancer Research
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, USA
- Department of Chemistry, Purdue University, West Lafayette, USA
- Center for Cancer Research
- Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
30
|
Tabata K, Saeki M, Yoshimori T, Hamasaki M. Monitoring and assessment of lysosomal membrane damage in cultured cells using the high-content imager. STAR Protoc 2023; 4:102236. [PMID: 37074905 PMCID: PMC10148077 DOI: 10.1016/j.xpro.2023.102236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/09/2023] [Accepted: 03/22/2023] [Indexed: 04/20/2023] Open
Abstract
Autophagy is an intracellular self-degradation process in which part of the cytoplasm, aggregates, or damaged organelles are degraded in lysosomes. Lysophagy is a specific form of selective autophagy responsible for clearing damaged lysosomes. Here, we present a protocol for inducing lysosomal damage in cultured cells and assessing lysosomal damage using a high-content imager and software program. We describe steps for induction of lysosomal damage, image acquisition with spinning disk confocal microscopy, and image analysis using Pathfinder. We then detail data analysis of the clearance of damaged lysosomes. For complete details on the use and execution of this protocol, please refer to Teranishi et al. (2022).1.
Collapse
Affiliation(s)
- Keisuke Tabata
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Bioscience, Osaka University, Osaka 565-0871, Japan; Department of Genetics, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Marika Saeki
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Bioscience, Osaka University, Osaka 565-0871, Japan
| | - Tamotsu Yoshimori
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Bioscience, Osaka University, Osaka 565-0871, Japan; Department of Genetics, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan
| | - Maho Hamasaki
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Bioscience, Osaka University, Osaka 565-0871, Japan; Department of Genetics, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
31
|
Bhattacharya A, Mukherjee R, Kuncha SK, Brunstein ME, Rathore R, Junek S, Münch C, Dikic I. A lysosome membrane regeneration pathway depends on TBC1D15 and autophagic lysosomal reformation proteins. Nat Cell Biol 2023; 25:685-698. [PMID: 37024685 DOI: 10.1038/s41556-023-01125-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023]
Abstract
Acute lysosomal membrane damage reduces the cellular population of functional lysosomes. However, these damaged lysosomes have a remarkable recovery potential independent of lysosomal biogenesis and remain unaffected in cells depleted in TFEB and TFE3. We combined proximity-labelling-based proteomics, biochemistry and high-resolution microscopy to unravel a lysosomal membrane regeneration pathway that depends on ATG8, the lysosomal membrane protein LIMP2, the RAB7 GTPase-activating protein TBC1D15 and proteins required for autophagic lysosomal reformation, including dynamin-2, kinesin-5B and clathrin. Following lysosomal damage, LIMP2 acts as a lysophagy receptor to bind ATG8, which in turn recruits TBC1D15 to damaged membranes. TBC1D15 interacts with ATG8 proteins on damaged lysosomes and provides a scaffold to assemble and stabilize the autophagic lysosomal reformation machinery. This potentiates the formation of lysosomal tubules and subsequent dynamin-2-dependent scission. TBC1D15-mediated lysosome regeneration was also observed in a cell culture model of oxalate nephropathy.
Collapse
Affiliation(s)
- Anshu Bhattacharya
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - Rukmini Mukherjee
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
- Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Santosh Kumar Kuncha
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | | | - Rajeshwari Rathore
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Stephan Junek
- Max Planck Institute of Biophysics, Frankfurt, Germany
- Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Christian Münch
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany.
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany.
- Max Planck Institute of Biophysics, Frankfurt, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany.
| |
Collapse
|
32
|
Lv X, Tang W, Qin J, Wang W, Dong J, Wei Y. The crosslinks between ferroptosis and autophagy in asthma. Front Immunol 2023; 14:1140791. [PMID: 37063888 PMCID: PMC10090423 DOI: 10.3389/fimmu.2023.1140791] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Autophagy is an evolutionarily conserved cellular process capable of degrading various biological molecules and organelles via the lysosomal pathway. Ferroptosis is a type of oxidative stress-dependent regulated cell death associated with the iron accumulation and lipid peroxidation. The crosslinks between ferroptosis and autophagy have been focused on since the dependence of ferroptosis on autophagy was discovered. Although the research and theories on the relationship between autophagy and ferroptosis remain scattered and fragmented, the crosslinks between these two forms of regulated cell death are closely related to the treatment of various diseases. Thereof, asthma as a chronic inflammatory disease has a tight connection with the occurrence of ferroptosis and autophagy since the crosslinked signal pathways may be the crucial regulators or exactly regulated by cells and secretion in the immune system. In addition, non-immune cells associated with asthma are also closely related to autophagy and ferroptosis. Further studies of cross-linking asthma inflammation with crosslinked signaling pathways may provide us with several key molecules that regulate asthma through specific regulators. The crosslinks between autophagy and ferroptosis provide us with a new perspective to interpret and understand the manifestations of asthma, potential drug discovery targets, and new therapeutic options to effectively intervene in the imbalance caused by abnormal inflammation in asthma. Herein, we introduce the main molecular mechanisms of ferroptosis, autophagy, and asthma, describe the role of crosslinks between ferroptosis and autophagy in asthma based on their common regulatory cells or molecules, and discuss potential drug discovery targets and therapeutic applications in the context of immunomodulatory and symptom alleviation.
Collapse
Affiliation(s)
- Xiaodi Lv
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingjing Qin
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wenqian Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
- *Correspondence: Ying Wei, ; Jingcheng Dong,
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
- *Correspondence: Ying Wei, ; Jingcheng Dong,
| |
Collapse
|
33
|
Vargas JNS, Hamasaki M, Kawabata T, Youle RJ, Yoshimori T. The mechanisms and roles of selective autophagy in mammals. Nat Rev Mol Cell Biol 2023; 24:167-185. [PMID: 36302887 DOI: 10.1038/s41580-022-00542-2] [Citation(s) in RCA: 475] [Impact Index Per Article: 237.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 11/09/2022]
Abstract
Autophagy is a process that targets various intracellular elements for degradation. Autophagy can be non-selective - associated with the indiscriminate engulfment of cytosolic components - occurring in response to nutrient starvation and is commonly referred to as bulk autophagy. By contrast, selective autophagy degrades specific targets, such as damaged organelles (mitophagy, lysophagy, ER-phagy, ribophagy), aggregated proteins (aggrephagy) or invading bacteria (xenophagy), thereby being importantly involved in cellular quality control. Hence, not surprisingly, aberrant selective autophagy has been associated with various human pathologies, prominently including neurodegeneration and infection. In recent years, considerable progress has been made in understanding mechanisms governing selective cargo engulfment in mammals, including the identification of ubiquitin-dependent selective autophagy receptors such as p62, NBR1, OPTN and NDP52, which can bind cargo and ubiquitin simultaneously to initiate pathways leading to autophagy initiation and membrane recruitment. This progress opens the prospects for enhancing selective autophagy pathways to boost cellular quality control capabilities and alleviate pathology.
Collapse
Affiliation(s)
- Jose Norberto S Vargas
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Maho Hamasaki
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Richard J Youle
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
34
|
Gong X, Wang Y, Tang Y, Wang Y, Zhang M, Li M, Zhang Y, Pan L. ATG16L1 adopts a dual-binding site mode to interact with WIPI2b in autophagy. SCIENCE ADVANCES 2023; 9:eadf0824. [PMID: 36857448 PMCID: PMC9977175 DOI: 10.1126/sciadv.adf0824] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Macroautophagy plays crucial roles in the regulation of cellular physiology and requires de novo synthesis of double-membrane autophagosomes, which relies on a specific interaction between autophagy-related 16L1 (ATG16L1) and WD repeat domain phosphoinositide-interacting protein 2b (WIPI2b). However, the molecular mechanism governing the interaction of ATG16L1 with WIPI2b remains elusive. Here, we find that ATG16L1 has two distinct binding sites for interacting with WIPI2b, the previously reported WIPI2b-binding site (WBS1) and the previously unidentified site (WBS2). We determine the crystal structures of WIPI2b with ATG16L1 WBS1 and WBS2, respectively, and elucidate the molecular mechanism underpinning the recruitment of ATG16L1 by WIPI2b. Moreover, we uncover that ATG16L1 WBS2 and its binding mode with WIPI2b is well conserved from yeast to mammals, unlike ATG16L1 WBS1. Last, our cell-based functional assays demonstrate that both ATG16L1 WBS1 and WBS2 are required for the effective autophagic flux. In conclusion, our findings provide mechanistic insights into the key ATG16L1/WIPI2b interaction in autophagy.
Collapse
Affiliation(s)
- Xinyu Gong
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yingli Wang
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yubin Tang
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yaru Wang
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Mingfang Zhang
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Miao Li
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, Hangzhou 310024, China
| | - Yuchao Zhang
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Lifeng Pan
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, Hangzhou 310024, China
| |
Collapse
|
35
|
Gallagher ER, Holzbaur ELF. The selective autophagy adaptor p62/SQSTM1 forms phase condensates regulated by HSP27 that facilitate the clearance of damaged lysosomes via lysophagy. Cell Rep 2023; 42:112037. [PMID: 36701233 PMCID: PMC10366342 DOI: 10.1016/j.celrep.2023.112037] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/16/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
In response to lysosomal damage, cells engage several quality-control mechanisms, including the selective isolation and degradation of damaged lysosomes by lysophagy. Here, we report that the selective autophagy adaptor SQSTM1/p62 is recruited to damaged lysosomes in both HeLa cells and neurons and is required for lysophagic flux. The Phox and Bem1p (PB1) domain of p62 mediates oligomerization and is specifically required for lysophagy. Consistent with this observation, we find that p62 forms condensates on damaged lysosomes. These condensates are precisely tuned by the small heat shock protein HSP27, which is phosphorylated in response to lysosomal injury and maintains the liquidity of p62 condensates, facilitating autophagosome formation. Mutations in p62 have been identified in patients with amyotrophic lateral sclerosis (ALS); ALS-associated mutations in p62 impair lysophagy, suggesting that deficits in this pathway may contribute to neurodegeneration. Thus, p62 condensates regulated by HSP27 promote lysophagy by forming platforms for autophagosome biogenesis at damaged lysosomes.
Collapse
Affiliation(s)
- Elizabeth R Gallagher
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Tsuchiya M, Kong W, Hiraoka Y, Haraguchi T, Ogawa H. TBK1 inhibitors enhance transfection efficiency by suppressing p62/SQSTM1 phosphorylation. Genes Cells 2023; 28:68-77. [PMID: 36284367 DOI: 10.1111/gtc.12987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 01/19/2023]
Abstract
DNA transfection is an essential technique in the life sciences. Non-viral transfection reagents are widely used for transfection in basic science. However, low transfection efficiency is a problem in some cell types. This low efficiency can be primarily attributed to the intracellular degradation of transfected DNA by p62-dependent selective autophagy, specifically by p62 phosphorylated at the S403 residue (p62-S403-P). To achieve efficient DNA transfection, we focused on a phosphorylation process that generates p62-S403-P and investigated whether inhibition of this process affects transfection efficiency. One of the kinases that phosphorylate p62 is TBK1. The TBK1 gene depletion in murine embryonic fibroblast cells by genome editing caused a significant reduction or loss of p62-S405-P (equivalent to human S403-P) and enhanced transfection efficiency, suggesting that TBK1 is a major kinase that phosphorylates p62 at S403. Therefore, TBK1 is a viable target for drug treatment to increase transfection efficiency. Transfection efficiency was enhanced when cells were treated with one of the following TBK1 inhibitors BX795, MRT67307, or amlexanox. This effect was synergistically improved when the two inhibitors were used in combination. Our results indicate that TBK1 inhibitors enhanced transfection efficiency by suppressing p62 phosphorylation.
Collapse
Affiliation(s)
- Megumi Tsuchiya
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Weixia Kong
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Yasushi Hiraoka
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Tokuko Haraguchi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Hidesato Ogawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
37
|
Lee YJ, Kim JK, Jung CH, Kim YJ, Jung EJ, Lee SH, Choi HR, Son YS, Shim SM, Jeon SM, Choe JH, Lee SH, Whang J, Sohn KC, Hur GM, Kim HT, Yeom J, Jo EK, Kwon YT. Chemical modulation of SQSTM1/p62-mediated xenophagy that targets a broad range of pathogenic bacteria. Autophagy 2022; 18:2926-2945. [PMID: 35316156 PMCID: PMC9673928 DOI: 10.1080/15548627.2022.2054240] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
The N-degron pathway is a proteolytic system in which the N-terminal degrons (N-degrons) of proteins, such as arginine (Nt-Arg), induce the degradation of proteins and subcellular organelles via the ubiquitin-proteasome system (UPS) or macroautophagy/autophagy-lysosome system (hereafter autophagy). Here, we developed the chemical mimics of the N-degron Nt-Arg as a pharmaceutical means to induce targeted degradation of intracellular bacteria via autophagy, such as Salmonella enterica serovar Typhimurium (S. Typhimurium), Escherichia coli, and Streptococcus pyogenes as well as Mycobacterium tuberculosis (Mtb). Upon binding the ZZ domain of the autophagic cargo receptor SQSTM1/p62 (sequestosome 1), these chemicals induced the biogenesis and recruitment of autophagic membranes to intracellular bacteria via SQSTM1, leading to lysosomal degradation. The antimicrobial efficacy was independent of rapamycin-modulated core autophagic pathways and synergistic with the reduced production of inflammatory cytokines. In mice, these drugs exhibited antimicrobial efficacy for S. Typhimurium, Bacillus Calmette-Guérin (BCG), and Mtb as well as multidrug-resistant Mtb and inhibited the production of inflammatory cytokines. This dual mode of action in xenophagy and inflammation significantly protected mice from inflammatory lesions in the lungs and other tissues caused by all the tested bacterial strains. Our results suggest that the N-degron pathway provides a therapeutic target in host-directed therapeutics for a broad range of drug-resistant intracellular pathogens.Abbreviations: ATG: autophagy-related gene; BCG: Bacillus Calmette-Guérin; BMDMs: bone marrow-derived macrophages; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CFUs: colony-forming units; CXCL: C-X-C motif chemokine ligand; EGFP: enhanced green fluorescent protein; IL1B/IL-1β: interleukin 1 beta; IL6: interleukin 6; LIR: MAP1LC3/LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; Mtb: Mycobacterium tuberculosis; MTOR: mechanistic target of rapamycin kinase; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PB1: Phox and Bem1; SQSTM1/p62: sequestosome 1; S. Typhimurium: Salmonella enterica serovar Typhimurium; TAX1BP1: Tax1 binding protein 1; TNF: tumor necrosis factor; UBA: ubiquitin-associated.
Collapse
Affiliation(s)
- Yoon Jee Lee
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Chan Hoon Jung
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eui Jung Jung
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Su Hyun Lee
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ha Rim Choi
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yeon Sung Son
- Neuroscience Research Institute, Medical Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sang Mi Shim
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sang Min Jeon
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jin Ho Choe
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC) & Basic Research Section, The Korean Institute of Tuberculosis (KIT), Cheongju, Korea
| | - Kyung-Cheol Sohn
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Department of Pharmacology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Gang Min Hur
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Department of Pharmacology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyun Tae Kim
- Chemistry R&D Center, AUTOTAC Bio Inc, Seoul, Republic of Korea
| | - Jinki Yeom
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea,Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea,CONTACT Eun-Kyeong Jo Department of Microbiology, and Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon35015, Korea
| | - Yong Tae Kwon
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea,Chemistry R&D Center, AUTOTAC Bio Inc, Seoul, Republic of Korea,SNU Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea,Yong Tae Kwon Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul110-799, Korea
| |
Collapse
|
38
|
Jia J, Wang F, Bhujabal Z, Peters R, Mudd M, Duque T, Allers L, Javed R, Salemi M, Behrends C, Phinney B, Johansen T, Deretic V. Stress granules and mTOR are regulated by membrane atg8ylation during lysosomal damage. J Cell Biol 2022; 221:e202207091. [PMID: 36179369 PMCID: PMC9533235 DOI: 10.1083/jcb.202207091] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 01/22/2023] Open
Abstract
We report that lysosomal damage is a hitherto unknown inducer of stress granule (SG) formation and that the process termed membrane atg8ylation coordinates SG formation with mTOR inactivation during lysosomal stress. SGs were induced by lysosome-damaging agents including SARS-CoV-2ORF3a, Mycobacterium tuberculosis, and proteopathic tau. During damage, mammalian ATG8s directly interacted with the core SG proteins NUFIP2 and G3BP1. Atg8ylation was needed for their recruitment to damaged lysosomes independently of SG condensates whereupon NUFIP2 contributed to mTOR inactivation via the Ragulator-RagA/B complex. Thus, cells employ membrane atg8ylation to control and coordinate SG and mTOR responses to lysosomal damage.
Collapse
Affiliation(s)
- Jingyue Jia
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Fulong Wang
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Zambarlal Bhujabal
- Autophagy Research Group, Institute of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ryan Peters
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Michal Mudd
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Thabata Duque
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Ruheena Javed
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Michelle Salemi
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, Davis, CA
| | - Christian Behrends
- Munich Cluster of Systems Neurology, Ludwig-Maximilians-Universität München, München, Germany
| | - Brett Phinney
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, Davis, CA
| | - Terje Johansen
- Autophagy Research Group, Institute of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
Teranishi H, Tabata K, Saeki M, Umemoto T, Hatta T, Otomo T, Yamamoto K, Natsume T, Yoshimori T, Hamasaki M. Identification of CUL4A-DDB1-WDFY1 as an E3 ubiquitin ligase complex involved in initiation of lysophagy. Cell Rep 2022; 40:111349. [PMID: 36103833 DOI: 10.1016/j.celrep.2022.111349] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/14/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023] Open
Abstract
Macroautophagy is a bulk degradation system in which double membrane-bound structures called autophagosomes to deliver cytosolic materials to lysosomes. Autophagy promotes cellular homeostasis by selectively recognizing and sequestering specific targets, such as damaged organelles, protein aggregates, and invading bacteria, termed selective autophagy. We previously reported a type of selective autophagy, lysophagy, which helps clear damaged lysosomes. Damaged lysosomes become ubiquitinated and recruit autophagic machinery. Proteomic studies using transfection reagent-coated beads and further evaluations reveal that a CUL4A-DDB1-WDFY1 E3 ubiquitin ligase complex is essential to initiate lysophagy and clear damaged lysosomes. Moreover, we show that LAMP2 is ubiquitinated by the CUL4A E3 ligase complex as a substrate on damaged lysosomes. These results reveal how cells selectively tag damaged lysosomes to initiate autophagy for the clearance of lysosomes.
Collapse
Affiliation(s)
- Hirofumi Teranishi
- JT Pharmaceutical Frontier Research Laboratory, Yokohama 236-0004, Japan
| | - Keisuke Tabata
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka 565-0871, Japan; Department of Genetics, Graduate School of Medicine Osaka University, Osaka 565-0871, Japan
| | - Marika Saeki
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka 565-0871, Japan
| | - Tetsuo Umemoto
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka 565-0871, Japan
| | - Tomohisa Hatta
- Molecular Profiling Research Center for Drug Discovery, AIST, Tokyo 135-0064, Japan
| | - Takanobu Otomo
- Department of Genetics, Graduate School of Medicine Osaka University, Osaka 565-0871, Japan
| | - Kentaro Yamamoto
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka 565-0871, Japan
| | - Toru Natsume
- Molecular Profiling Research Center for Drug Discovery, AIST, Tokyo 135-0064, Japan
| | - Tamotsu Yoshimori
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka 565-0871, Japan; Department of Genetics, Graduate School of Medicine Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan.
| | - Maho Hamasaki
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka 565-0871, Japan; Department of Genetics, Graduate School of Medicine Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
43
|
Kravić B, Bionda T, Siebert A, Gahlot P, Levantovsky S, Behrends C, Meyer H. Ubiquitin profiling of lysophagy identifies actin stabilizer CNN2 as a target of VCP/p97 and uncovers a link to HSPB1. Mol Cell 2022; 82:2633-2649.e7. [PMID: 35793674 DOI: 10.1016/j.molcel.2022.06.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/11/2022] [Accepted: 06/08/2022] [Indexed: 11/26/2022]
Abstract
Lysosomal membrane permeabilization (LMP) is an underlying feature of diverse conditions including neurodegeneration. Cells respond by extensive ubiquitylation of membrane-associated proteins for clearance of the organelle through lysophagy that is facilitated by the ubiquitin-directed AAA-ATPase VCP/p97. Here, we assessed the ubiquitylated proteome upon acute LMP and uncovered a large diversity of targets and lysophagy regulators. They include calponin-2 (CNN2) that, along with the Arp2/3 complex, translocates to damaged lysosomes and regulates actin filaments to drive phagophore formation. Importantly, CNN2 needs to be ubiquitylated during the process and removed by VCP/p97 for efficient lysophagy. Moreover, we identified the small heat shock protein HSPB1 that assists VCP/p97 in the extraction of CNN2 and show that other membrane regulators including SNAREs, PICALM, AGFG1, and ARL8B are ubiquitylated during lysophagy. Our data reveal a framework of how ubiquitylation and two effectors, VCP/p97 and HSPB1, cooperate to protect cells from the deleterious effects of LMP.
Collapse
Affiliation(s)
- Bojana Kravić
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Tihana Bionda
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Alexander Siebert
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Pinki Gahlot
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Sophie Levantovsky
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377 Munich, Germany.
| | - Hemmo Meyer
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
44
|
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
|
45
|
Borgo GM, Burke TP, Tran CJ, Lo NTN, Engström P, Welch MD. A patatin-like phospholipase mediates Rickettsia parkeri escape from host membranes. Nat Commun 2022; 13:3656. [PMID: 35760786 PMCID: PMC9237051 DOI: 10.1038/s41467-022-31351-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/15/2022] [Indexed: 12/25/2022] Open
Abstract
Rickettsia species of the spotted fever group are arthropod-borne obligate intracellular bacteria that can cause mild to severe human disease. These bacteria invade host cells, replicate in the cell cytosol, and spread from cell to cell. To access the host cytosol and avoid immune detection, they escape membrane-bound vacuoles by expressing factors that disrupt host membranes. Here, we show that a patatin-like phospholipase A2 enzyme (Pat1) facilitates Rickettsia parkeri infection by promoting escape from host membranes and cell-cell spread. Pat1 is important for infection in a mouse model and, at the cellular level, is crucial for efficiently escaping from single and double membrane-bound vacuoles into the host cytosol, and for avoiding host galectins that mark damaged membranes. Pat1 is also important for avoiding host polyubiquitin, preventing recruitment of autophagy receptor p62, and promoting actin-based motility and cell-cell spread. Pathogenic Rickettsia species are arthropod-borne, obligate intracellular bacteria that invade host cells, replicate in the cell cytosol, and spread from cell to cell. Here, Borgo et al. identify a Rickettsia phospholipase enzyme that is important for infection by helping the bacteria escape from host cell vacuoles into the host cytosol, preventing targeting by autophagy, and promoting bacterial motility and spread to other cells.
Collapse
Affiliation(s)
- Gina M Borgo
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Thomas P Burke
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.,Department of Microbiology & Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Cuong J Tran
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Nicholas T N Lo
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Patrik Engström
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.,Primordial Genetics, San Diego, CA, USA
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
46
|
The role of K63-linked polyubiquitin in several types of autophagy. Biol Futur 2022; 73:137-148. [DOI: 10.1007/s42977-022-00117-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/05/2022] [Indexed: 01/08/2023]
Abstract
AbstractLysosomal-dependent self-degradative (autophagic) mechanisms are essential for the maintenance of normal homeostasis in all eukaryotic cells. Several types of such self-degradative and recycling pathways have been identified, based on how the cellular self material can incorporate into the lysosomal lumen. Ubiquitination, a well-known and frequently occurred posttranslational modification has essential role in all cell biological processes, thus in autophagy too. The second most common type of polyubiquitin chain is the K63-linked polyubiquitin, which strongly connects to some self-degradative mechanisms in the cells. In this review, we discuss the role of this type of polyubiquitin pattern in numerous autophagic processes.
Collapse
|
47
|
Bikorimana JP, Salame N, Beaudoin S, Balood M, Crosson T, Abusarah J, Talbot S, Löbenberg R, Plouffe S, Rafei M. Promoting antigen escape from dendritic cell endosomes potentiates anti-tumoral immunity. Cell Rep Med 2022; 3:100534. [PMID: 35492876 PMCID: PMC9040180 DOI: 10.1016/j.xcrm.2022.100534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The cross-presenting capacity of dendritic cells (DCs) can be limited by non-specific degradation during endosome maturation. To bypass this limitation, we present in this study a new Accum-based formulation designed to promote endosome-to-cytosol escape. Treatment of primary DCs with Accum linked to the xenoantigen ovalbumin (OVA) triggers endosomal damages and enhances protein processing. Despite multiple challenges using ascending doses of tumor cells, DC prophylactic vaccination results in complete protection due to increased levels of effector CD4 and CD8 T cells as well as high production of pro-inflammatory mediators. When combined with anti-PD-1, therapeutic vaccination using both syngeneic and allogeneic Accum-OVA-pulsed DCs triggers potent anti-tumoral responses. The net outcome culminates in increased CD11c, CD8, and NK infiltration along with a high CD8/Treg ratio. These highly favorable therapeutic effects highlight the promising potential of Accum as a distinct and potent technology platform suitable for the design of next generation cell cancer vaccines. Accum-linked antigen enhances antigen processing and presentation Pulsed dendritic cells elicit potent effector T cell responses Therapeutic vaccination using allogeneic DCs controls pre-established tumors The vaccine boosts tumor-infiltrating lymphocytes and increases the CD8/Treg ratio
Collapse
Affiliation(s)
- Jean-Pierre Bikorimana
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, QC, Canada
| | - Natasha Salame
- Department of Biomedical Sciences, Université de Montréal, Montréal, QC, Canada
| | - Simon Beaudoin
- Research and Development Branch, Defence Therapeutics Inc., Vancouver, BC, Canada
| | - Mohammad Balood
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, Canada
| | - Théo Crosson
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, Canada
| | - Jamilah Abusarah
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, Canada
| | - Sebastien Talbot
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, Canada
| | - Raimar Löbenberg
- Department of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Sebastien Plouffe
- Research and Development Branch, Defence Therapeutics Inc., Vancouver, BC, Canada
| | - Moutih Rafei
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, QC, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, Canada.,Molecular Biology Program, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
48
|
Protocols to monitor TFEB activation following lysosomal damage in cultured cells using microscopy and immunoblotting. STAR Protoc 2022; 3:101018. [PMID: 35243365 PMCID: PMC8872846 DOI: 10.1016/j.xpro.2021.101018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Following lysosomal damage, activation and nuclear translocation of transcription factor EB (TFEB) is the key event to maintain lysosomal homeostasis. Here, we describe steps to induce lysosomal damage in HeLa cells. This can be followed by monitoring the changes in TFEB localization using widefield fluorescence microscopy. As a complementary approach, we describe the use of immunoblotting to follow the activation and localization of TFEB in cell lysates. These protocols enable quantitative analysis of TFEB. For complete details on the use and execution of this protocol, please refer to Nakamura et al. (2020). Detailed protocol to induce lysosomal damage in cultured cells Microscopy-based analysis of changes in TFEB localization in response to damage Immunoblotting approach to follow the activation and localization of TFEB in cell lysates
Collapse
|
49
|
Zoncu R, Perera RM. Built to last: lysosome remodeling and repair in health and disease. Trends Cell Biol 2022; 32:597-610. [DOI: 10.1016/j.tcb.2021.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/21/2022]
|
50
|
Date Y, Matsuura A, Itakura E. Disruption of actin dynamics induces autophagy of the eukaryotic chaperonin TRiC/CCT. Cell Death Dis 2022; 8:37. [PMID: 35079001 PMCID: PMC8789831 DOI: 10.1038/s41420-022-00828-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/11/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022]
Abstract
Autophagy plays important role in the intracellular protein quality control system by degrading abnormal organelles and proteins, including large protein complexes such as ribosomes. The eukaryotic chaperonin tailless complex polypeptide 1 (TCP1) ring complex (TRiC), also called chaperonin-containing TCP1 (CCT), is a 1-MDa hetero-oligomer complex comprising 16 subunits that facilitates the folding of ~10% of the cellular proteome that contains actin. However, the quality control mechanism of TRiC remains unclear. To monitor the autophagic degradation of TRiC, we generated TCP1α-RFP-GFP knock-in HeLa cells using a CRISPR/Cas9-knock-in system with an RFP-GFP donor vector. We analyzed the autophagic degradation of TRiC under several stress conditions and found that treatment with actin (de)polymerization inhibitors increased the lysosomal degradation of TRiC, which was localized in lysosomes and suppressed by deficiency of autophagy-related genes. Furthermore, we found that treatment with actin (de)polymerization inhibitors increased the association between TRiC and unfolded actin, suggesting that TRiC was inactivated. Moreover, unfolded actin mutants were degraded by autophagy. Taken together, our results indicate that autophagy eliminates inactivated TRiC, serving as a quality control system.
Collapse
|