1
|
Küng C, Lazarou M, Nguyen TN. Advances in mitophagy initiation mechanisms. Curr Opin Cell Biol 2025; 94:102493. [PMID: 40117675 DOI: 10.1016/j.ceb.2025.102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/23/2025]
Abstract
Mitophagy is an important lysosomal degradative pathway that removes damaged or unwanted mitochondria to maintain cellular and organismal homeostasis. The mechanisms behind how mitophagy is initiated to form autophagosomes around mitochondria have gained a lot of interest since they can be potentially targeted by mitophagy-inducing therapeutics. Mitophagy initiation can be driven by various autophagy receptors or adaptors that respond to different cellular and mitochondrial stimuli, ranging from mitochondrial damage to metabolic rewiring. This review will cover recent advances in our understanding of how mitophagy is initiated, and by doing so reveal the mechanistic plasticity of how autophagosome formation can begin.
Collapse
Affiliation(s)
- Catharina Küng
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Lazarou
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD 20185, USA.
| | - Thanh Ngoc Nguyen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD 20185, USA.
| |
Collapse
|
2
|
Cai J, Zhou H, Liu M, Zhang D, Lv J, Xue H, Zhou H, Zhang W. Host immunity and intracellular bacteria evasion mechanisms: Enhancing host-directed therapies with drug delivery systems. Int J Antimicrob Agents 2025; 65:107492. [PMID: 40107461 DOI: 10.1016/j.ijantimicag.2025.107492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Host-directed therapies (HDTs) have been investigated as a potential solution to combat intracellular and drug-resistant bacteria. HDTs stem from extensive research on the intricate interactions between the host and intracellular bacteria, leading to a treatment approach that relies on immunoregulation. To improve the bioavailability and safety of HDTs, researchers have utilized diverse drug delivery systems (DDS) to encapsulate and transport therapeutic agents to target cells. In this review, we first introduce the three mechanisms of bactericidal action and intracellular bacterial evasion: autophagy, reactive oxygen species (ROS), and inflammatory cytokines, with a particular focus on autophagy. Special attention is given to the detailed mechanism of xenophagy in clearing intracellular bacteria, a crucial selective autophagy process that specifically targets and degrades intracellular pathogens. Following this, we present the application of DDS to modulate these regulatory methods for intracellular bacteria elimination. By integrating insights from immunology and nanomedicine, this review highlights the emerging role of DDS in advancing HDTs for intracellular bacterial infections and paving the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Jiayang Cai
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Han Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Mingwei Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Dingjian Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Jingxuan Lv
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Haokun Xue
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Houcheng Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
3
|
Deng L, Wang J, Liu Y, Wang L, Liu F, Lou B, Zhu J. Hypoxia-induced mitochondrial dysfunction and mitophagy in the small yellow croaker (Larimichthys polyactis). FISH & SHELLFISH IMMUNOLOGY 2025; 161:110275. [PMID: 40086497 DOI: 10.1016/j.fsi.2025.110275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/18/2025] [Accepted: 03/12/2025] [Indexed: 03/16/2025]
Abstract
Mitophagy serves as a pivotal mechanism for regulating the quantity and quality of mitochondria within cells, exerting significant influence on various processes such as cell differentiation, oxidative stress, inflammatory responses, and apoptosis. Currently, research on whether and how fish activate mitophagy under hypoxic stress conditions is still insufficient. In this study, to determine the mechanisms whereby marine fish adapt to hypoxic environments from the perspective of mitophagy, we used the small yellow croaker (Larimichthys polyactis) as the research subject and combined in vivo (liver) and in vitro (small yellow croaker fry [SYCF] cell line) hypoxic stress experiments. Fish exposed to hypoxic conditions were found to be characterized by liver tissue damage, and we detected significant elevations in the levels of hydrogen peroxide in liver tissues and reactive oxygen species (ROS) in SYCF cells, along with significant reductions in mitochondrial membrane potential. These findings thus indicate that hypoxic stress leads to tissue damage, excessive ROS production, and mitochondrial damage. In further experiments, we pre-treated SYCF cells with the antioxidant N-acetylcysteine, which was found to effectively reduce ROS levels and prevented the loss of mitochondrial membrane potential, thereby indicating that ROS play a crucial role in hypoxic stress-induced mitochondrial damage. Subsequently, to investigate whether hypoxic stress activates mitophagy to remove damaged mitochondria, we examined changes in the mRNA expression of mitophagy-related genes (bnip3, lc3b, bnip3l, beclin1, fundc1, and ulk1) in the liver and SYCF cells of L. polyactis exposed to hypoxic stress, and detected a significant upregulation of the mRNA expression of these genes. Furthermore, examination of liver ultrastructure and changes in the co-localization of mitochondria and lysosomes in SYCF cells revealed that hypoxic stress induces the formation of autophagosomes and autolysosomes in the liver, with an enhanced co-localization of mitochondria and lysosomes being observed after 6 h of hypoxia, which gradually increased with a prolongation of hypoxic exposure. We have, for the first time, exhibited the formation process of autophagosomes and the subsequent formation of autolysosomes in fish under hypoxic stress. These findings reveal the induction of mitophagy in L. polyactis in response to hypoxic stress, and indicate that these fish may initiate a mitophagic response to remove damaged mitochondria, reduce excessive ROS accumulation, and maintain cellular homeostasis. Our findings will not only lay a biological foundation for the breeding of hypoxia-tolerant strains of L. polyactis but also provide new insights into the mechanisms underlying the adaptation of marine fish to hypoxic environments.
Collapse
Affiliation(s)
- Lu Deng
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Jingqian Wang
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Yang Liu
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Li Wang
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Feng Liu
- Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province, 310021, People's Republic of China
| | - Bao Lou
- Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province, 310021, People's Republic of China.
| | - Junquan Zhu
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China.
| |
Collapse
|
4
|
Barros D, Ferreira BH, Garcia-Gonzalez P, Carbone F, Luka M, Leite-Pinheiro F, Machado MD, Nikolaou T, Pilotti A, Goguet E, Antas P, Mendes A, Zhang L, Cresci M, Galliot L, Gigan JP, Reverendo M, Su B, Narita M, Paton AW, Paton JC, Rocchi S, Rieux-Laucat F, Argüello RJ, Nal B, Liang Y, Ménager M, Gatti E, Almeida CR, Pierre P. Induction of the ISR by AB5 subtilase cytotoxin drives type-I IFN expression in pDCs via STING activation. Proc Natl Acad Sci U S A 2025; 122:e2421258122. [PMID: 40388626 DOI: 10.1073/pnas.2421258122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
We demonstrate that exposure to the AB5 subtilase cytotoxin (SubAB) induces the unfolded protein response (UPR) in human peripheral blood mononuclear cells, concomitant with a proinflammatory response across distinct cell subsets. Notably, SubAB selectively induces type-I interferon (IFN) expression in plasmacytoid dendritic cells, acting synergistically with Toll-like receptor 7 stimulation. The induction of type-I IFN in response to SubAB relies on stimulator of interferon genes (STING) activation, coupled with protein synthesis inhibition mediated by protein kinase R-like endoplasmic reticulum kinase (PERK) and phosphorylation of the eukaryotic translation initiation factor 2 subunit-alpha. By impeding mRNA translation through the integrated stress response, SubAB precipitates the downregulation of the negative innate signaling feedback regulator Tax1-binding protein 1. This downregulation is necessary to unleash TANK-binding kinase 1 signaling associated with STING activation. These findings shed light on how UPR-inducing conditions may regulate the immune system during infection or pathogenesis.
Collapse
Affiliation(s)
- Daniela Barros
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Beatriz H Ferreira
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Paulina Garcia-Gonzalez
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Francesco Carbone
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Marine Luka
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Fátima Leite-Pinheiro
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Mariana D Machado
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Theopisti Nikolaou
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Angelo Pilotti
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Eliot Goguet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Paulo Antas
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Andreia Mendes
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Lichen Zhang
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Marina Cresci
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Lou Galliot
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Julien P Gigan
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Marisa Reverendo
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Bing Su
- Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Miwako Narita
- Laboratory of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata 951-8518, Japan
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Frédéric Rieux-Laucat
- Université de Paris Cité, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris 75015, France
| | - Rafael J Argüello
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Béatrice Nal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Yinming Liang
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Mickaël Ménager
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Evelina Gatti
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Catarina R Almeida
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Philippe Pierre
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| |
Collapse
|
5
|
Yang X, Lv L, Zhang Y, Zhang Z, Zeng S, Zhang X, Wang Q, Dorf M, Li S, Fu B. ATP2A2 regulates STING1/MITA-driven signal transduction including selective autophagy. Autophagy 2025:1-16. [PMID: 40265346 DOI: 10.1080/15548627.2025.2496786] [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: 11/25/2024] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025] Open
Abstract
STING1/MITA not only induces innate immune responses but also triggers macroautophagy/autophagy to selectively degrade signaling molecules. However, the molecular mechanisms regulating STING1-mediated selective autophagy remain unclear. Here, we first report that ATP2A2 directly interacts with STING1, regulating STING1-mediated innate immune response by modulating its polymerization and trafficking, thereby inhibiting DNA virus infection. Notably, while screening for reticulophagy receptors involved in STING1-mediated selective autophagy, we identified SEC62 as an important receptor protein in STING1-mediated reticulophagy. Mechanistically, SEC62 strengthens its interaction with STING1 upon activation and concurrently facilitates STING1-mediated reticulophagy upon starvation, which are dependent on ATP2A2. Furthermore, knocking down SEC62 in WT cells inhibits STING1-mediated MAP1LC3B/LC3B lipidation and autophagosome formation, an effect that is lost in ATP2A2 knockout cells, suggesting that SEC62's role in STING1-mediated selective autophagy is ATP2A2 dependent. Thus, our findings identify the reticulophagy receptor SEC62 as a novel receptor protein regulating STING1-mediated selective autophagy, providing new insight into the mechanism regarding a reticulophagy receptor in the process of STING1-induced selective autophagy.Abbrevations: aa: amino acids; AP-MS: affinity tag purification-mass spectrometry; ATP2A1: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 1; ATP2A2: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2; ATP2A3: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 3; CANX: calnexin; CCPG1: cell cycle progression 1; CGAS: cyclic GMP-AMP synthase; ctDNA: calf thymus DNA; dsRNA: double-stranded RNA; diABZI: diamidobenzimidazole; ER: endoplasmic reticulum; ERGIC: ER-Golgi intermediate compartment; EBSS: Earle's Balanced Salt Solution; EV: empty vector; FL: full length; GOLGA2/GM130: golgin A2; HSV-1: herpes simplex virus type 1; IRF3: interferon regulatory factor 3; IFNs: type I interferons; ISD: interferon stimulatory DNA; KO: knockout; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; poly(I:C): polyinosinic-polycytidylic acid; NBR1: NBR1 autophagy cargo receptor; PRR: pattern recognition receptor; reticulophagy: selective autophagic degradation of the ER; RETREG1/FAM134B: reticulophagy regulator 1; RIGI: RNA sensor RIG-I; RTN3L: reticulon 3; SEC62: SEC62 homolog, preprotein translocation factor; SeV: Sendai virus; STIM1: stromal interaction molecule 1; STING1/MITA: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1; TEX264: testis expressed 264, ER-phagy receptor; TMX1: thioredoxin related transmembrane protein 1; VSV: vesicular stomatitis virus; VACV: vaccinia virus; ZMPSTE24: zinc metallopeptidase STE24.
Collapse
Affiliation(s)
- Xue Yang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Linyue Lv
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yuelan Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Zhuyou Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Shaowei Zeng
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xinyi Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Qinyang Wang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Martin Dorf
- Department of Microbiology & Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Shitao Li
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, USA
| | - Bishi Fu
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Diaz PJ, Shi Q, McNeish PY, Banerjee S. Tubulin Polymerization Promoting Proteins: Functional Diversity With Implications in Neurological Disorders. J Neurosci Res 2025; 103:e70044. [PMID: 40317801 PMCID: PMC12047068 DOI: 10.1002/jnr.70044] [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: 01/30/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Tubulin Polymerization Promoting Proteins (TPPPs) are highly conserved across species but remain poorly understood. There are three TPPP genes in humans, with only one homologous TPPP gene in invertebrates, such as Drosophila and C. elegans. The human TPPP (TPPP1/p25/p25α) is enriched in the brain and shares sequence similarities with the invertebrate TPPPs. TPPP/p25 associates with microtubules and plays a pivotal role in microtubule dynamics, bundling, and polymerization, thereby stabilizing the microtubular network. This is essential for cytoskeletal organization and proper functioning of neurons and glial cells, including axonal growth, regeneration, migration, trafficking, synapse formation, and myelination of axons. However, studies have also uncovered that besides its cytoplasmic/microtubular localization, TPPP/p25 is present in other subcellular compartments, including the mitochondria and nucleus, underscoring the presence of additional novel functions. At the molecular level, TPPP/p25 is predicted to exist as an intrinsically disordered protein and is implicated in neurological and neurodegenerative disorders, including Parkinson's and related disorders and Multiple Sclerosis. In this article, we provide a comprehensive overview of TPPP/p25, highlighting its evolutionary conservation, cellular and subcellular localization, established and emerging functions in the nervous system, interacting partners, potential clinical relevance to human neurological disorders, and conclude with unresolved questions and future areas of study.
Collapse
Affiliation(s)
- Paloma J. Diaz
- Department of Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio, Joe R. and Teresa Lozano Long School of MedicineSan AntonioTexasUSA
| | - Qian Shi
- Department of Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio, Joe R. and Teresa Lozano Long School of MedicineSan AntonioTexasUSA
- Center for Biomedical NeuroscienceUniversity of Texas Health Science Center San Antonio, Joe R. and Teresa Lozano Long School of MedicineSan AntonioTexasUSA
| | - Priscilla Y. McNeish
- Department of Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio, Joe R. and Teresa Lozano Long School of MedicineSan AntonioTexasUSA
| | - Swati Banerjee
- Department of Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio, Joe R. and Teresa Lozano Long School of MedicineSan AntonioTexasUSA
- Center for Biomedical NeuroscienceUniversity of Texas Health Science Center San Antonio, Joe R. and Teresa Lozano Long School of MedicineSan AntonioTexasUSA
- Perry and Ruby Stevens Parkinson's Disease Center of ExcellenceUniversity of Texas Health Science Center San Antonio, Joe R. and Teresa Lozano Long School of MedicineSan AntonioTexasUSA
| |
Collapse
|
7
|
Ananphongmanee V, Tungwaravut S, Yingsunthonwattana W, Sangsuriya P, Tassanakajon A, Somboonwiwat K. Shrimp autophagy receptor protein PvTAX1BP1 regulates autophagy and facilitates white spot syndrome virus replication in Penaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110386. [PMID: 40315936 DOI: 10.1016/j.fsi.2025.110386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/20/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
Tax1 binding protein 1 (TAX1BP1) plays a role in autophagy regulation and proteasomal pathway across different species, though its function in shrimp is still being explored. In Penaeus vannamei, the homolog PvTAX1BP1 was characterized by a CALCOCO1 domain, a LC3-interacting region (LIR), two coiled-coil domains, and two ubiquitin-binding zinc finger regions (UBZs). It was ubiquitously expressed in shrimp tissues while its expression in hemocytes was notably downregulated following white spot syndrome virus (WSSV) infection. Silencing PvTAX1BP1 reduced WSSV replication and prolonged shrimp survival, suggesting its involvement in viral pathogenesis. Immunofluorescence assay revealed co-localization of PvTAX1BP1 with the autophagy-related protein PvLC3, indicating a potential interaction and its role in LC3-mediated autophagy. Additionally, knockdown of PvTAX1BP1 resulted in downregulation of the autophagy marker PvLC3-II in WSSV-infected shrimp, reinforcing its role in autophagy regulation during infection. Both yeast two-hybrid (Y2H) and immunofluorescence assays confirmed that PvTAX1BP1 directly interacts with three WSSV proteins: WSSV325, WSSV458, and WSSV517. These findings suggest that PvTAX1BP1 facilitates WSSV replication by modulating host LC3-mediated autophagy, potentially through its interactions with WSSV proteins. This highlights a mechanism by which viruses can exploit cellular processes for their own benefit.
Collapse
Affiliation(s)
- Vorawit Ananphongmanee
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phayathai Rd., Bangkok, 10330, Thailand
| | - Suttipong Tungwaravut
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phayathai Rd., Bangkok, 10330, Thailand
| | - Warumporn Yingsunthonwattana
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phayathai Rd., Bangkok, 10330, Thailand
| | - Pakkakul Sangsuriya
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, 12120, Thailand
| | - Anchalee Tassanakajon
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phayathai Rd., Bangkok, 10330, Thailand
| | - Kunlaya Somboonwiwat
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phayathai Rd., Bangkok, 10330, Thailand.
| |
Collapse
|
8
|
Che S, Zhang Y, Xu H, Shi J, Hou Y. TBB inhibits CK2/PD-L1/EGFR pathway-mediated tumor progression. Eur J Pharmacol 2025; 999:177689. [PMID: 40311835 DOI: 10.1016/j.ejphar.2025.177689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
The expression of PD-L1 on cancer cells facilitates tumor immune escape by binding to PD-1 on T cells, thereby inhibiting T cell activity. However, the role of intracellular PD-L1 signaling in tumor progression remains unclear. In this study, we demonstrate that CK2 induces PD-L1 phosphorylation at Thr-285, which enhances PD-L1 protein stability. This phosphorylation disrupts the interaction between LC3B and PD-L1, inhibiting PD-L1 degradation via autophagy. Furthermore, PD-L1-T285 phosphorylation promotes EGFR binding to PD-L1, leading to activation of EGFR downstream signaling. This activation drives non-small cell lung cancer (NSCLC) cell proliferation, migration, invasion, and tumor growth. Conversely, CK2 depletion or treatment with a CK2 inhibitor reversed these effects. Our findings reveal a novel mechanism by which the CK2/PD-L1/EGFR pathway promotes tumor progression.
Collapse
Affiliation(s)
- Suning Che
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Yao Zhang
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Huihui Xu
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Juanjuan Shi
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Yongzhong Hou
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China.
| |
Collapse
|
9
|
Minami A, Nishi K, Yamada R, Jinnai G, Shima H, Oishi S, Akagawa H, Aono T, Hidaka M, Masaki H, Kuzuyama T, Noda Y, Ogawa T. The ribonuclease RNase T2 mediates selective autophagy of ribosomes induced by starvation in Saccharomyces cerevisiae. J Biol Chem 2025; 301:108554. [PMID: 40294649 DOI: 10.1016/j.jbc.2025.108554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
RNase T2 is a conserved ribonuclease, playing essential and diverse roles despite its simple enzymatic activity. Saccharomyces cerevisiae RNase T2, known as Rny1p, is stress-responsive and localizes in the vacuole. Upon starvation, ribosomes are degraded by autophagy, in which Rny1p mediates rRNA degradation. However, whether the ribosomal degradation is selective or nonselective is still being determined in S. cerevisiae. Here, we elucidated novel aspects of ribosome degradation mechanisms and the function of Rny1p in stress response. We discovered that most ribosomes are selectively degraded, whose mechanism differs from the previously reported selective degradation process called "ribophagy." Rsa1p, a factor involved in assembling 60S ribosomal subunits, is revealed to interact with Atg8p and act as a receptor for selective ribosome degradation in the cytosol. The accumulation of rRNA in vacuoles, due to lack of Rny1p, leads to a decrease in nonselective autophagic activity. This is one of the reasons for the inability of Rny1p-deficient strains to adapt to starvation conditions. Rny1p is also reported to be secreted and associated with the cell wall. We revealed that a C-terminal extension of Rny1p, characteristic in some fungal RNase T2, is required to anchor the cell wall. Some nonfungal RNase T2 proteins also have C-terminal extensions. However, their sequences and structures differ from those of fungal RNase T2, suggesting that their biological functions may also be distinct. The diversity of C-terminal extensions across different organisms is thought to be one reason why RNase T2 plays various roles.
Collapse
Affiliation(s)
- Atsushi Minami
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kohei Nishi
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Rikusui Yamada
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Gai Jinnai
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hikari Shima
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sakiko Oishi
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hirofumi Akagawa
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Toshihiro Aono
- Agro-Biotechnology Research Center (AgTECH), The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Makoto Hidaka
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Haruhiko Masaki
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomohisa Kuzuyama
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Collaborative Research Institute for Innovative Microbiology (CRIIM), The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yoichi Noda
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Collaborative Research Institute for Innovative Microbiology (CRIIM), The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tetsuhiro Ogawa
- Department of Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Collaborative Research Institute for Innovative Microbiology (CRIIM), The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
10
|
Bi J, Sun Y, Guo M, Sun X, Sun J, Jiang R, Wang N, Huang G. Lysosomes: guardians and healers within cells- multifaceted perspective and outlook from injury repair to disease treatment. Cancer Cell Int 2025; 25:136. [PMID: 40205430 PMCID: PMC11984033 DOI: 10.1186/s12935-025-03771-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Lysosomes, as crucial organelles within cells, carry out diverse biological functions such as waste degradation, regulation of the cellular environment, and precise control of cell signaling. This paper reviews the core functions and structural characteristics of lysosomes, and delves into the current research status of lysosomes damage repair mechanisms. Subsequently, we explore in depth the close association between lysosomes and various diseases, including but not limited to age-related chronic diseases, neuro-degenerative diseases, tumors, inflammation, and immune imbalance. Additionally, we also provide a detailed discussion of the application of lysosome-targeted substances in the field of regenerative medicine, especially the enormous potential demonstrated in key areas such as stem cell regulation and therapy, and myocardial cell repair. Though the integration of multidisciplinary research efforts, we believe that lysosomes damage repair mechanisms will demonstrate even greater application value in disease treatment and regenerative medicine.
Collapse
Affiliation(s)
- Jianlei Bi
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China
| | - Yincong Sun
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Meihua Guo
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Xiaoxin Sun
- College of Integrative Medicine, Dalian Medical University, Dalian, 116044, Liaoning, P.R. China
| | - Jie Sun
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Rujiao Jiang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Gena Huang
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China.
| |
Collapse
|
11
|
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
|
12
|
Han S, Zhao X, Yu C, Cui C, Zhang Y, Zhu Q, Qiu M, Yang C, Yin H. Nestin Regulates Autophagy-Dependent Ferroptosis Mediated Skeletal Muscle Atrophy by Ubiquitinating MAP 1LC3B. J Cachexia Sarcopenia Muscle 2025; 16:e13779. [PMID: 40183241 PMCID: PMC11969254 DOI: 10.1002/jcsm.13779] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Programmed cell death plays a critical role in skeletal muscle atrophy. Ferroptosis, an iron-dependent form of programmed cell death driven by lipid peroxidation, has been implicated in various diseases, but its role in skeletal muscle atrophy remains unclear. METHODS Ferroptosis in skeletal muscle atrophy was investigated using two models: dexamethasone (Dex)-induced atrophy (n = 6 independent cell cultures per group) and simulated microgravity (n = 6 mice per group). Conditional Nestin knockout (KO) mice were generated using CRISPR/Cas9 (n = 6-8 mice per group), with wild-type (WT) controls (n = 6-8). Phenotypic analyses included histopathology (HE staining), functional assessments (muscle strength, weight analysis, treadmill), and dystrophy evaluation (dystrophin staining). Molecular analyses involved flow cytometry, ELISA, transmission electron microscopy, PI staining, and IP/MS to delineate Nestin-regulated ferroptosis pathways in skeletal muscle atrophy. RESULTS Ferroptosis was significantly activated in both atrophy models, with a 2.5-fold increase in lipid peroxidation (p < 0.01), a 2-fold accumulation of Fe2+ (p < 0.01) and a 50% reduction in Nestin expression (p < 0.001). Nestin KO mice exhibited exacerbated muscle atrophy, showing a 40% decrease in muscle weight (p < 0.01) and a 30% reduction in muscle strength (p < 0.05) compared to WT mice. Nestin overexpression mitigated Dex-induced ferroptosis, reducing lipid peroxidation by 40%, decreasing Fe2+ accumulation by 50% (p < 0.01), and improving muscle function by 30% (p < 0.05). Mechanistically, Nestin interacted with MAP 1LC3B (LC3B) to catalyse LC3B polyubiquitination at lysine-51, reducing LC3B availability for autophagy and inhibiting autophagy flux by 60% (p < 0.01), leading to a 50% reduction in ferroptosis (p < 0.001). CONCLUSIONS Our study identifies Nestin as a critical regulator of ferroptosis-autophagy crosstalk in skeletal muscle atrophy. Targeting Nestin-LC3B ubiquitination may offer novel therapeutic strategies for preventing muscle wasting in diseases such as cachexia and sarcopenia.
Collapse
Affiliation(s)
- Shunshun Han
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Xiyu Zhao
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Chunlin Yu
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Can Cui
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Yao Zhang
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Qing Zhu
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Mohan Qiu
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Huadong Yin
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| |
Collapse
|
13
|
Saar D, Lennartsson CLE, Weidner P, Burgermeister E, Kragelund BB. The Myotubularin Related Proteins and the Untapped Interaction Potential of Their Disordered C-Terminal Regions. Proteins 2025; 93:831-854. [PMID: 39614773 PMCID: PMC11878207 DOI: 10.1002/prot.26774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 03/05/2025]
Abstract
Intrinsically disordered regions (IDRs) of proteins remain understudied with enigmatic sequence features relevant to their functions. Members of the myotubularin-related protein (MTMR) family contain uncharacterized IDRs. After decades of research on their phosphatase activity, recent work on the C-terminal IDRs of MTMR7 revealed new interactions and important new functions beyond the phosphatase function. Here we take a broader look at the C-terminal domains (CTDs) of 14 human MTMRs and use bioinformatic tools and biophysical methods to ask which other functions may be probable in this protein family. The predictions show that the CTDs are disordered and carry short linear motifs (SLiMs) important for targeting of MTMRs to defined subcellular compartments and implicating them in signaling, phase separation, interaction with diverse proteins, including transcription factors and are of relevance for cancer research and neuroscience. We also present experimental methods to study the CTDs and use them to characterize the coiled coil (CC) domains of MTMR7 and MTMR9. We show homo- and hetero-oligomerization with preference for MTMR7-CC to form dimers, while MTMR9-CC forms trimers. We relate the results to sequence features and make predictions for the structural landscape of other MTMRs. Our work gives a broad insight into the so far unrecognized features and SLiMs in MTMR-CTDs, and provides the basis for more in-depth experimental research on this diverse protein family and understudied IDRs in proteins in general.
Collapse
Affiliation(s)
- Daniel Saar
- REPINUniversity of CopenhagenCopenhagenDenmark
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Structural Biology and NMR Laboratory, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | | | - Philip Weidner
- Department of Medicine II, University Medical Center MannheimHeidelberg UniversityMannheimGermany
| | - Elke Burgermeister
- Department of Medicine II, University Medical Center MannheimHeidelberg UniversityMannheimGermany
| | - Birthe B. Kragelund
- REPINUniversity of CopenhagenCopenhagenDenmark
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Structural Biology and NMR Laboratory, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
14
|
Marinković M, Rožić A, Polančec D, Novak I. Cost-effective and simple flow cytometry quantification of receptor-mediated autophagy using fluorescent tagging. FEBS Open Bio 2025; 15:587-598. [PMID: 39716041 PMCID: PMC11961372 DOI: 10.1002/2211-5463.13958] [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: 08/06/2024] [Revised: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024] Open
Abstract
Mitophagy, a selective clearance of damaged or superfluous mitochondria via autophagy machinery and lysosomal degradation, is an evolutionarily conserved process essential for various physiological functions, including cellular differentiation and immune responses. Defects in mitophagy are implicated in numerous human diseases, such as neurodegenerative disorders, cancer, and metabolic conditions. Despite significant advancements in mitophagy research over recent decades, novel and robust methodologies are necessary to elucidate its molecular mechanisms comprehensively. In this study, we present a detailed protocol for quantitatively assessing mitophagy through flow cytometry using a mitochondria-targeted fluorescent mitophagy receptor, GFP-BNIP3L/NIX. This method offers a rapid alternative to conventional microscopy or immunoblotting techniques for analyzing mitophagy activity. Additionally, this approach can theoretically be adapted to utilize any fluorescent-tagged selective autophagy receptor, enabling the direct and rapid analysis of various types of receptor-mediated selective autophagy.
Collapse
Affiliation(s)
| | - Ana Rožić
- School of MedicineUniversity of SplitCroatia
| | | | - Ivana Novak
- School of MedicineUniversity of SplitCroatia
| |
Collapse
|
15
|
Feng C, Hu Z, Zhao M, Leng C, Li G, Yang F, Fan X. Region-specific mitophagy in nucleus pulposus, annulus fibrosus, and cartilage endplate of intervertebral disc degeneration: mechanisms and therapeutic strategies. Front Pharmacol 2025; 16:1579507. [PMID: 40248091 PMCID: PMC12003974 DOI: 10.3389/fphar.2025.1579507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent condition contributing to various spinal disorders, posing a significant global health burden. Mitophagy plays a crucial role in maintaining mitochondrial quantity and quality and is closely associated with the onset and progression of IVDD. Well-documented region-specific mitophagy mechanisms in IVDD are guiding the development of therapeutic strategies. In the nucleus pulposus (NP), impaired mitochondria lead to apoptosis, oxidative stress, senescence, extracellular matrix degradation and synthesis, excessive autophagy, inflammation, mitochondrial instability, and pyroptosis, with key regulatory targets including AMPK, PGC-1α, SIRT1, SIRT3, Progerin, p65, Mfn2, FOXO3, NDUFA4L2, SLC39A7, ITGα5/β1, Nrf2, and NLRP3 inflammasome. In the annulus fibrosus (AF), mitochondrial damage induces apoptosis and oxidative stress mediated by PGC-1α, while in the cartilage endplate (CEP), mitochondrial dysfunction similarly triggers apoptosis and oxidative stress. These mechanistic insights highlight therapeutic strategies such as activating Parkin-dependent and Ub-independent mitophagy pathways for NP, enhancing Parkin-dependent mitophagy for AF, and targeting Parkin-mediated mitophagy for CEP. These strategies include the use of natural ingredients, hormonal modulation, gene editing technologies, targeted compounds, and manipulation of related proteins. This review summarizes the mechanisms of mitophagy in different regions of the intervertebral disc and highlights therapeutic approaches using mitophagy modulators to ameliorate IVDD. It discusses the complex mechanisms of mitophagy and underscores its potential as a therapeutic target. The objective is to provide valuable insights and a scientific basis for the development of mitochondrial-targeted drugs for anti-IVDD.
Collapse
Affiliation(s)
- Chaoqun Feng
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziang Hu
- Department of Orthopedics, The TCM Hospital of Longquanyi District, Chengdu, China
| | - Min Zhao
- International Ward (Gynecology), Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Leng
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangye Li
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Yang
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Fan
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Zhang S, Luo X, Yuan X, Wu D, Liu J, Zhao K, Xu Y, Zhou J, Li X, Li QX. Crystal Structure of Autophagy-Associated Protein 8 at 1.36 Å Resolution and Its Inhibitory Interactions with Indole Analogs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7111-7120. [PMID: 40066832 PMCID: PMC11951139 DOI: 10.1021/acs.jafc.4c11205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
Autophagy-associated protein 8 (ATG8) is essential for autophagy and organismal growth and development. In this study, we successfully resolved the crystal structure of Drosophila melanogaster (D. melanogaster) ATG8a (DmATG8a) at 1.36 Å resolution. Being distinct from previously characterized ATG8 homologues, DmATG8a (121 residues) adopts a unique fold comprising five α-helices and four β-folding strands, in contrast to the canonical four α-helices and four β-folding strands observed in other ATG8 proteins. DmATG8a features two active cavities: hydrophobic pocket 1 (HP1) and hydrophobic pocket 2 (HP2), which are essential for the normal physiological function of ATG8. Indole and its analogs can bind specifically with HP1. Microscale thermophoresis results demonstrated a strong affinity of 6-fluoroindole with DmATG8a (3.54 μmol/L), but no affinity with the DmATG8aK48A mutant, suggesting that Lys48 is critical in binding 6-fluoroindole probably via a hydrogen bond interaction. The half-maximum lethal concentration (LC50) of 6-fluoroindole against D. melanogaster adult flies was 169 μg/mL. Our findings establish DmATG8a as a promising target for developing indole-based insecticides.
Collapse
Affiliation(s)
- Shanqi Zhang
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Xin Luo
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Xiu Yuan
- Department
of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu 96822, Hawaii United States
| | - Danxia Wu
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Jing Liu
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Kunhong Zhao
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Youwei Xu
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Jingjiang Zhou
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Xiangyang Li
- State Key
Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and
Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Qing X. Li
- Department
of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu 96822, Hawaii United States
- Hawaii Pacific
Neuroscience, 2230 Liliha
Street, Honolulu 96817, Hawaii, United States
| |
Collapse
|
17
|
Bouvier C, Gonzalez-Santamarta M, Profitós-Pelejà N, Armengol M, Quinet G, Alasseur Q, Ceccato L, Xolalpa W, Freire R, Guillermet-Guibert J, Reybier K, Caminade AM, Beck HC, Carvalho AS, Matthiesen R, Rain JC, Sutherland JD, Barrio R, Roué G, Rodriguez MS. Role of TRIM24 in the regulation of proteasome-autophagy crosstalk in bortezomib-resistant mantle cell lymphoma. Cell Death Discov 2025; 11:108. [PMID: 40097385 PMCID: PMC11914149 DOI: 10.1038/s41420-025-02355-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/07/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Resistance to bortezomib (BTZ) represents a major bottleneck to continue using this proteasome inhibitor in the treatment of mantle cell lymphoma (MCL). In this study, we investigated the mechanisms by which TRIM24 (tripartite motif-containing 24), a ubiquitin ligase enriched in the ubiquitome of BTZ-resistant MCL cells, modulates proteasome-autophagy crosstalk. The localization and stability of TRIM24 were differentially influenced by the inhibition of proteasome or autophagy in MCL cells with acquired BTZ resistance (ZBR). Moreover, genetic deletion of the TRIM24 gene in ZBR (ZBRTRIM24 KO) effectively impaired cell proliferation without impacting the degradation of the proteasome by proteaphagy that is typically observed in BTZ-resistant cells. Notably, pre-treatment of ZBR cells with a proteolysis-targeting chimera (PROTAC) targeting TRIM24 (dTRIM24) successfully restored BTZ susceptibility, underscoring the critical role of TRIM24 in mediating resistance to proteasome inhibition. Interestingly, the combined apoptogenic activity of dTRIM24 and BTZ was preserved in a second BTZ-resistant clone (JBR) that lacks functional p53, indicating that this tumor suppressor is not required for the observed effect. Furthermore, we demonstrated that reducing TRIM24 protein levels in BTZ-resistant cells via dTRIM24 treatment restored proteasome activity, facilitating efficient apoptosis induction in cells exposed to the dTRIM24/BTZ combination. Mechanistically, dTRIM24 treatment promoted the formation of K48-linked ubiquitin chains and their association with proteasome subunits, specifically in BTZ-resistant cells. Taken together, these findings reveal that TRIM24 plays a pivotal regulatory role in the crosstalk between the proteasome and autophagy in BTZ-resistant MCL cells by modulating ubiquitin chain abundance, thereby influencing the activation of one or the other proteolytic pathway.
Collapse
Affiliation(s)
- Corentin Bouvier
- Laboratoire de Chimie de Coordination (LCC) CNRS-UPR8241, Toulouse, 31077, France
| | | | - Núria Profitós-Pelejà
- Lymphoma Translational Group, UBIRed, Josep Carreras Leukaemia Research Institute, 08916, Badalona, Spain
| | - Marc Armengol
- Lymphoma Translational Group, UBIRed, Josep Carreras Leukaemia Research Institute, 08916, Badalona, Spain
| | - Grégoire Quinet
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Investigación Sanitaria de Canarias (IISC), La Laguna, La Laguna, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, 38200, Santa Cruz de Tenerife, Spain
| | | | - Laurie Ceccato
- Laboratoire de Chimie de Coordination (LCC) CNRS-UPR8241, Toulouse, 31077, France
| | - Wendy Xolalpa
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, UNAM, 62210, Cuernavaca, Morelos, Mexico
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Investigación Sanitaria de Canarias (IISC), La Laguna, La Laguna, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, 38200, Santa Cruz de Tenerife, Spain
- Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Julie Guillermet-Guibert
- Centre de Recherche en Cancerologie de Toulouse (CRCT), Inserm, CNRS, Université de Toulouse, Toulouse, 31100, France
| | - Karine Reybier
- PharmaDev, UMR 152, Université de Toulouse, IRD, UT3, 31400, Toulouse, France
| | - Anne-Marie Caminade
- Laboratoire de Chimie de Coordination (LCC) CNRS-UPR8241, Toulouse, 31077, France
| | - Hans C Beck
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, iNOVA4Health, Nova Medical School, Facultade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082, Lisboa, Portugal
| | - Rune Matthiesen
- Computational and Experimental Biology Group, iNOVA4Health, Nova Medical School, Facultade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082, Lisboa, Portugal
| | | | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Gaël Roué
- Lymphoma Translational Group, UBIRed, Josep Carreras Leukaemia Research Institute, 08916, Badalona, Spain.
| | - Manuel S Rodriguez
- Laboratoire de Chimie de Coordination (LCC) CNRS-UPR8241, Toulouse, 31077, France.
- BMolecular, Centre Pierre Potier, Toulouse, 31100, France.
- PharmaDev, UMR 152, Université de Toulouse, IRD, UT3, 31400, Toulouse, France.
| |
Collapse
|
18
|
Hao Y, Li R, Fan C, Gao Y, Hou X, wen W, Shen Y. Identification and validation of mitophagy-related genes in acute myocardial infarction and ischemic cardiomyopathy and study of immune mechanisms across different risk groups. Front Immunol 2025; 16:1486961. [PMID: 40114920 PMCID: PMC11922711 DOI: 10.3389/fimmu.2025.1486961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Acute myocardial infarction (AMI) is a critical condition that can lead to ischemic cardiomyopathy (ICM), a subsequent heart failure state characterized by compromised cardiac function. Methods This study investigates the role of mitophagy in the transition from AMI to ICM. We analyzed AMI and ICM datasets from GEO, identifying mitophagy-related differentially expressed genes (MRDEGs) through databases like GeneCards and Molecular Signatures Database, followed by functional enrichment and Protein-Protein Interaction analyses. Logistic regression, Support Vector Machine, and LASSO (Least Absolute Shrinkage and Selection Operator) were employed to pinpoint key MRDEGs and develop diagnostic models, with risk stratification performed using LASSO scores. Subgroup analyses included functional enrichment and immune infiltration analysis, along with protein domain predictions and the integration of regulatory networks involving Transcription Factors, miRNAs, and RNA-Binding Proteins, leading to drug target identification. Results The TGFβ pathway showed significant differences between high- and low-risk groups in AMI and ICM. Notably, in the AMI low-risk group, MRDEGs correlated positively with activated CD4+ T cells and negatively with Type 17 T helper cells, while in the AMI high-risk group, RPS11 showed a positive correlation with natural killer cells. In ICM, MRPS5 demonstrated a negative correlation with activated CD4+ T cells in the low-risk group and with memory B cells, mast cells, and dendritic cells in the high-risk group. The diagnostic accuracy of RPS11 was validated with an area under the curve (AUC) of 0.794 across diverse experimental approaches including blood samples, animal models, and myocardial hypoxia/reoxygenation models. Conclusions This study underscores the critical role of mitophagy in the transition from AMI to ICM, highlighting RPS11 as a highly significant biomarker with promising diagnostic potential and therapeutic implications.
Collapse
Affiliation(s)
- Ying Hao
- Department of Cardiovascular Medicine, State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiovascular Medicine, Shanghai East Hospital Ji’an Hospital, Ji’an, Jiangxi, China
| | - RuiLin Li
- Department of Cardiovascular Medicine, State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiovascular Medicine, Shanghai East Hospital Ji’an Hospital, Ji’an, Jiangxi, China
| | - ChengHui Fan
- Department of Cardiovascular Medicine, State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Cardiovascular Medicine, Shanghai East Hospital Ji’an Hospital, Ji’an, Jiangxi, China
| | - Yang Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xia Hou
- Department of Cardiovascular Medicine, State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei wen
- Department of Cardiovascular Medicine, State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - YunLi Shen
- Department of Cardiovascular Medicine, State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
19
|
Yang L, You J, Yang X, Jiao R, Xu J, Zhang Y, Mi W, Zhu L, Ye Y, Ren R, Min D, Tang M, Chen L, Li F, Liu P. ACSS2 drives senescence-associated secretory phenotype by limiting purine biosynthesis through PAICS acetylation. Nat Commun 2025; 16:2071. [PMID: 40021646 PMCID: PMC11871226 DOI: 10.1038/s41467-025-57334-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Senescence-associated secretory phenotype (SASP) mediates the biological effects of senescent cells on the tissue microenvironment and contributes to ageing-associated disease progression. ACSS2 produces acetyl-CoA from acetate and epigenetically controls gene expression through histone acetylation under various circumstances. However, whether and how ACSS2 regulates cellular senescence remains unclear. Here, we show that pharmacological inhibition and deletion of Acss2 in mice blunts SASP and abrogates the pro-tumorigenic and immune surveillance functions of senescent cells. Mechanistically, ACSS2 directly interacts with and promotes the acetylation of PAICS, a key enzyme for purine biosynthesis. The acetylation of PAICS promotes autophagy-mediated degradation of PAICS to limit purine metabolism and reduces dNTP pools for DNA repair, exacerbating cytoplasmic chromatin fragment accumulation and SASP. Altogether, our work links ACSS2-mediated local acetyl-CoA generation to purine metabolism through PAICS acetylation that dictates the functionality of SASP, and identifies ACSS2 as a potential senomorphic target to prevent senescence-associated diseases.
Collapse
Affiliation(s)
- Li Yang
- Research and Innovation Center, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Jianwei You
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Xincheng Yang
- Research and Innovation Center, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Ruishu Jiao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Xu
- Research and Innovation Center, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Yue Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Wen Mi
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Lingzhi Zhu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Youqiong Ye
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruobing Ren
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Delin Min
- Research and Innovation Center, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Meilin Tang
- Research and Innovation Center, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Li Chen
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Fuming Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Pingyu Liu
- Research and Innovation Center, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
20
|
Huang X, Zhang J, Yao J, Mi N, Yang A. Phase separation of p62: roles and regulations in autophagy. Trends Cell Biol 2025:S0962-8924(25)00033-9. [PMID: 40011090 DOI: 10.1016/j.tcb.2025.01.010] [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: 10/06/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/28/2025]
Abstract
The phase separation of the cargo receptor sequestome-1/p62 (SQSTM1/p62) is a critical mechanism for assembling signaling complexes in autophagy. During this process, p62 undergoes phase separation upon binding to polyubiquitin chains, concentrating ubiquitinated substrates within p62 droplets. These droplets further gather membrane sources and core autophagy machineries to facilitate autophagosome formation. The dynamics of p62 droplets are finely tuned in response to autophagy signals triggered by cellular stresses. Recent studies have revealed new regulatory mechanisms that highlight the significance of p62 phase separation in regulating autophagy. This review summarizes and discusses the molecular mechanisms of p62 phase separation and its roles in autophagy, with particular emphasis on the regulation of p62 droplets and their interaction modes with autophagic membranes.
Collapse
Affiliation(s)
- Xue Huang
- School of Life Sciences, Chongqing University, Chongqing 401331, China; Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Jinpei Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China; Basic Medical College, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China; Key Laboratory of High Incidence Disease Research in Xinjiang (Xinjiang Medical University), Ministry of Education, Urumqi, 830011, Xinjiang, China
| | - Jia Yao
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Na Mi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China; Basic Medical College, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China; Key Laboratory of High Incidence Disease Research in Xinjiang (Xinjiang Medical University), Ministry of Education, Urumqi, 830011, Xinjiang, China.
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
21
|
Huang R, Han B, Peng J, Jiao H. PTB Regulates Keloid Fibroblast Migration and Proliferation Through Autophagy. Aesthetic Plast Surg 2025; 49:897-907. [PMID: 39402202 DOI: 10.1007/s00266-024-04375-6] [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: 07/16/2024] [Accepted: 09/03/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Keloid disease is a chronic fibroproliferative disease that occurs after tissue injury, and the currently available treatments are unsatisfactory. OBJECTIVES We aimed to explore the level of autophagy in keloid fibroblasts (KFbs) and adjacent normal fibroblasts (NFbs). In addition, whether polypyrimidine tract-binding protein (PTB) regulates the biological functions of KFbs via autophagy was also investigated. METHODS The morphology of fibroblasts in normal skin and keloids was observed transmission electron microscopy. We silenced PTB with PTB-specific siRNA to determine whether PTB-regulated KFb proliferation. Acridine orange and LysoTracker Red staining was performed to label acidic compartments. Interestingly, when autophagy was inhibited by wortmannin, the PTB knockdown-mediated decrease in KFb migration and proliferation was abolished, while the collagen I and III levels were not altered; these results indicated that PTB regulated the migration and proliferation of KFbs via autophagy, while collagen synthesis occurred independently of PTB regulation. RESULTS Many activities related to the survival and function of KFbs are controlled by PTB. Transmission electron microscopy revealed more autophagosomes and autolysosomes in KFbs than in NFbs. PTB induced autophagy in KFbs, as demonstrated by the significantly greater number of autophagosomes in KFbs after PTB knockdown, which was revealed by acridine orange and LysoTracker staining. CONCLUSIONS Our study is the first to show that PTB regulates the migration and proliferation of KFbs via autophagy and that PTB regulates collagen synthesis in KFbs in an autophagy-independent manner. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Rong Huang
- Department of Dermatology, Hangzhou Third People's Hospital, 38, West Lake Avenue, Hangzhou, 310009, Zhejiang, China
| | - Bing Han
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Ba-Da-Chu Road, Beijing, 100144, China
| | - Jianzhong Peng
- Department of Dermatology, Hangzhou Third People's Hospital, 38, West Lake Avenue, Hangzhou, 310009, Zhejiang, China.
| | - Hu Jiao
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Ba-Da-Chu Road, Beijing, 100144, China.
| |
Collapse
|
22
|
Gou Q, Yan B, Duan Y, Guo Y, Qian J, Shi J, Hou Y. Ubiquitination of CD47 Regulates Innate Anti-Tumor Immune Response. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412205. [PMID: 39665172 PMCID: PMC11792004 DOI: 10.1002/advs.202412205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/16/2024] [Indexed: 12/13/2024]
Abstract
In addition to adaptive immune checkpoint of PD-1/PD-L1, the innate immune checkpoint SIRPα/CD47 plays an important role in regulation of tumor immune escape. However, the mechanism of CD47 ubiquitination on tumor immune escape remains unclear. Here it is found that TRAF2 bound to the C-terminal of CD47 cytoplasmic fragment and induced its ubiquitination, leading to inhibition of CD47 autophagic degradation by disrupting its binding to LC3, which in turn inhibited macrophage phagocytosis and promoted tumor immune escape. In contrast, loss of TRAF2 facilitated CD47 autophagic degradation and inhibited tumor immune escape. Moreover, autophagy induction promoted CD47 degradation and enhanced the efficacy of CD47 antibody anti-tumor immunotherapy. These findings revealed a novel mechanism of ubiquitination of CD47 on tumor immune escape.
Collapse
Affiliation(s)
- Qian Gou
- School of Life ScienceJiangsu UniversityZhenjiangJiangsu Province212013People's Republic of China
| | - Bingjun Yan
- School of Life ScienceJiangsu UniversityZhenjiangJiangsu Province212013People's Republic of China
| | - Yalan Duan
- School of Life ScienceJiangsu UniversityZhenjiangJiangsu Province212013People's Republic of China
| | - Yilei Guo
- School of Life ScienceJiangsu UniversityZhenjiangJiangsu Province212013People's Republic of China
| | - Jing Qian
- School of Life ScienceJiangsu UniversityZhenjiangJiangsu Province212013People's Republic of China
| | - Juanjuan Shi
- School of Life ScienceJiangsu UniversityZhenjiangJiangsu Province212013People's Republic of China
| | - Yongzhong Hou
- School of Life ScienceJiangsu UniversityZhenjiangJiangsu Province212013People's Republic of China
| |
Collapse
|
23
|
Markitantova Y, Simirskii V. Retinal Pigment Epithelium Under Oxidative Stress: Chaperoning Autophagy and Beyond. Int J Mol Sci 2025; 26:1193. [PMID: 39940964 PMCID: PMC11818496 DOI: 10.3390/ijms26031193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
The structural and functional integrity of the retinal pigment epithelium (RPE) plays a key role in the normal functioning of the visual system. RPE cells are characterized by an efficient system of photoreceptor outer segment phagocytosis, high metabolic activity, and risk of oxidative damage. RPE dysfunction is a common pathological feature in various retinal diseases. Dysregulation of RPE cell proteostasis and redox homeostasis is accompanied by increased reactive oxygen species generation during the impairment of phagocytosis, lysosomal and mitochondrial failure, and an accumulation of waste lipidic and protein aggregates. They are the inducers of RPE dysfunction and can trigger specific pathways of cell death. Autophagy serves as important mechanism in the endogenous defense system, controlling RPE homeostasis and survival under normal conditions and cellular responses under stress conditions through the degradation of intracellular components. Impairment of the autophagy process itself can result in cell death. In this review, we summarize the classical types of oxidative stress-induced autophagy in the RPE with an emphasis on autophagy mediated by molecular chaperones. Heat shock proteins, which represent hubs connecting the life supporting pathways of RPE cells, play a special role in these mechanisms. Regulation of oxidative stress-counteracting autophagy is an essential strategy for protecting the RPE against pathological damage when preventing retinal degenerative disease progression.
Collapse
Affiliation(s)
- Yuliya Markitantova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | | |
Collapse
|
24
|
Aponte-Diaz D, Harris JM, Kang TE, Korboukh V, Sotoudegan MS, Gray JL, Yennawar NH, Moustafa IM, Macadam A, Cameron CE. Non-lytic spread of poliovirus requires the nonstructural protein 3CD. mBio 2025; 16:e0327624. [PMID: 39665531 PMCID: PMC11708018 DOI: 10.1128/mbio.03276-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Non-enveloped viruses like poliovirus (PV) have evolved the capacity to spread by non-lytic mechanisms. For PV, this mechanism exploits the host secretory autophagy pathway. Virions are selectively incorporated into autophagosomes, double-membrane vesicles that travel to the plasma membrane, fuse, and release single-membrane vesicles containing virions. Loading of cellular cargo into autophagosomes relies on direct or indirect interactions with microtubule-associated protein 1B-light chain 3 (LC3) that are mediated by motifs referred to as LC3-interaction regions (LIRs). We have identified a PV mutant with a severe defect in non-lytic spread. An F-to-Y substitution in a putative LIR of the nonstructural protein 3CD prevented virion incorporation into LC3-positive autophagosomes and virion trafficking to the plasma membrane for release. Using high-angle annular dark-field scanning transmission electron microscopy to monitor PV-induced autophagosome biogenesis, for the first time, we show that virus-induced autophagic signals yield normal autophagosomes, even in the absence of virions. The F-to-Y derivative of PV 3CD was unable to support normal autophagosome biogenesis. Together, these studies make a compelling case for the direct role of a viral nonstructural protein in the formation and loading of the vesicular carriers used for non-lytic spread that may depend on the proper structure, accessibility, and/or dynamics of its LIR. The studies of PV 3CD protein reported here will hopefully provoke a more deliberate look at the presence and function of LIR motifs in viral proteins of viruses known to use autophagy as the basis for non-lytic spread. IMPORTANCE Poliovirus (PV) and other enteroviruses hijack the cellular secretory autophagy pathway for non-lytic virus transmission. While much is known about the cellular factors required for non-lytic transmission, much less is known about viral factors contributing to transmission. We have discovered a PV nonstructural protein required for multiple steps of the pathway leading to vesicle-enclosed virions. This discovery should facilitate the identification of the specific steps of the cellular secretory autophagy pathway and corresponding factors commandeered by the virus and may uncover novel targets for antiviral therapy.
Collapse
Affiliation(s)
- David Aponte-Diaz
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jayden M. Harris
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tongjia Ella Kang
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Victoria Korboukh
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Mohamad S. Sotoudegan
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer L. Gray
- Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Neela H. Yennawar
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Ibrahim M. Moustafa
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Andrew Macadam
- Division of Vaccines, Medicines and Healthcare Products Regulatory Agency, Potters Bar, Herts., United Kingdom
| | - Craig E. Cameron
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
25
|
Xu Q, Wang H, Yang R, Tao Y, Wang Z, Zhang S, Sun B, Li D, Lu B, Liu C. α-Synuclein amyloid fibril directly binds to LC3B and suppresses SQSTM1/p62-mediated selective autophagy. Cell Res 2025; 35:72-75. [PMID: 39300253 PMCID: PMC11701113 DOI: 10.1038/s41422-024-01022-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- Qianhui Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huilan Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, New Cornerstone Science Laboratory, School of Life Sciences, Fudan University, Shanghai, China
| | - Ruonan Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, New Cornerstone Science Laboratory, School of Life Sciences, Fudan University, Shanghai, China
| | - Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Ziying Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, New Cornerstone Science Laboratory, School of Life Sciences, Fudan University, Shanghai, China
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Bo Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Boxun Lu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, New Cornerstone Science Laboratory, School of Life Sciences, Fudan University, Shanghai, China.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
26
|
Panda M, Markaki M, Tavernarakis N. Mitostasis in age-associated neurodegeneration. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167547. [PMID: 39437856 DOI: 10.1016/j.bbadis.2024.167547] [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: 04/06/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Mitochondria are essential organelles that play crucial roles in various metabolic and signalling pathways. Proper neuronal function is highly dependent on the health of these organelles. Of note, the intricate structure of neurons poses a critical challenge for the transport and distribution of mitochondria to specific energy-intensive domains, such as synapses and dendritic appendages. When faced with chronic metabolic challenges and bioenergetic deficits, neurons undergo degeneration. Unsurprisingly, disruption of mitostasis, the process of maintaining cellular mitochondrial content and function within physiological limits, has been implicated in the pathogenesis of several age-associated neurodegenerative disorders. Indeed, compromised integrity and metabolic activity of mitochondria is a principal hallmark of neurodegeneration. In this review, we survey recent findings elucidating the role of impaired mitochondrial homeostasis and metabolism in the onset and progression of age-related neurodegenerative disorders. We also discuss the importance of neuronal mitostasis, with an emphasis on the major mitochondrial homeostatic and metabolic pathways that contribute to the proper functioning of neurons. A comprehensive delineation of these pathways is crucial for the development of early diagnostic and intervention approaches against neurodegeneration.
Collapse
Affiliation(s)
- Mrutyunjaya Panda
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece; Department of Medicine, University of Verona, Verona 37134, Italy; Faculdade de Farmácia, University of Lisbon, Lisbon 1649-003, Portugal
| | - Maria Markaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece; Division of Basic Sciences, School of Medicine, University of Crete, Heraklion 71003, Crete, Greece.
| |
Collapse
|
27
|
Herhaus L, Gestal-Mato U, Eapen VV, Mačinković I, Bailey HJ, Prieto-Garcia C, Misra M, Jacomin AC, Ammanath AV, Bagarić I, Michaelis J, Vollrath J, Bhaskara RM, Bündgen G, Covarrubias-Pinto A, Husnjak K, Zöller J, Gikandi A, Ribičić S, Bopp T, van der Heden van Noort GJ, Langer JD, Weigert A, Harper JW, Mancias JD, Dikic I. IRGQ-mediated autophagy in MHC class I quality control promotes tumor immune evasion. Cell 2024; 187:7285-7302.e29. [PMID: 39481378 DOI: 10.1016/j.cell.2024.09.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 06/24/2024] [Accepted: 09/29/2024] [Indexed: 11/02/2024]
Abstract
The autophagy-lysosome system directs the degradation of a wide variety of cargo and is also involved in tumor progression. Here, we show that the immunity-related GTPase family Q protein (IRGQ), an uncharacterized protein to date, acts in the quality control of major histocompatibility complex class I (MHC class I) molecules. IRGQ directs misfolded MHC class I toward lysosomal degradation through its binding mode to GABARAPL2 and LC3B. In the absence of IRGQ, free MHC class I heavy chains do not only accumulate in the cell but are also transported to the cell surface, thereby promoting an immune response. Mice and human patients suffering from hepatocellular carcinoma show improved survival rates with reduced IRGQ levels due to increased reactivity of CD8+ T cells toward IRGQ knockout tumor cells. Thus, we reveal IRGQ as a regulator of MHC class I quality control, mediating tumor immune evasion.
Collapse
Affiliation(s)
- Lina Herhaus
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Uxía Gestal-Mato
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Vinay V Eapen
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Institutes of Medicine, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Igor Mačinković
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Institute of Biochemistry I, Goethe University School of Medicine, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Henry J Bailey
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Riedberg Campus, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Cristian Prieto-Garcia
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Mohit Misra
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Riedberg Campus, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Anne-Claire Jacomin
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Aparna Viswanathan Ammanath
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ivan Bagarić
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jolina Michaelis
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Joshua Vollrath
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Riedberg Campus, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Goethe University Frankfurt, Riedberg Campus, 60438 Frankfurt am Main, Germany
| | - Ramachandra M Bhaskara
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Riedberg Campus, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Georg Bündgen
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Adriana Covarrubias-Pinto
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Koraljka Husnjak
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jonathan Zöller
- Max Planck Institute of Biophysics, Goethe University Frankfurt, Riedberg Campus, 60438 Frankfurt am Main, Germany
| | - Ajami Gikandi
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Institutes of Medicine, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Sara Ribičić
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Julian D Langer
- Max Planck Institute of Biophysics, Goethe University Frankfurt, Riedberg Campus, 60438 Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe University School of Medicine, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Joseph D Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Institutes of Medicine, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Ivan Dikic
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Riedberg Campus, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Goethe University Frankfurt, Riedberg Campus, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
28
|
Khalafiyan A, Fadaie M, Khara F, Zarrabi A, Moghadam F, Khanahmad H, Cordani M, Boshtam M. Highlighting roles of autophagy in human diseases: a perspective from single-cell RNA sequencing analyses. Drug Discov Today 2024; 29:104224. [PMID: 39521332 DOI: 10.1016/j.drudis.2024.104224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Autophagy, the lysosome-driven breakdown of intracellular components, is pivotal in regulating eukaryotic cellular processes and maintaining homeostasis, making it physiologically important even under normal conditions. Cellular mechanisms involving autophagy include the response to nutrient deprivation, intracellular quality control, early development, and cell differentiation. Despite its established health significance, the role of autophagy in cancer and other diseases remains complex and not fully understood. A comprehensive understanding of autophagy is crucial to facilitate the development of novel therapies and drugs that can protect and improve human health. High-throughput technologies, such as single-cell RNA sequencing (scRNA-seq), have enabled researchers to study transcriptional landscapes at single-cell resolution, significantly advancing our knowledge of autophagy pathways across diverse physiological and pathological contexts. This review discusses the latest advances in scRNA-seq for autophagy research and highlights its potential in the molecular characterization of various diseases.
Collapse
Affiliation(s)
- Anis Khalafiyan
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmood Fadaie
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Khara
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Fariborz Moghadam
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
29
|
Asghari N, Saei AK, Cordani M, Nayeri Z, Moosavi MA. Drug repositioning identifies potential autophagy inhibitors for the LIR motif p62/SQSTM1 protein. Comput Biol Chem 2024; 113:108235. [PMID: 39369612 DOI: 10.1016/j.compbiolchem.2024.108235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Autophagy is a critical cellular process for degrading damaged organelles and proteins under stressful conditions and has casually been shown to contribute to tumor survival and drug resistance. Sequestosome-1 (SQSTM1/p62) is an autophagy receptor that interacts with its binding partners via the LC3-interacting region (LIR). The p62 protein has been a highly researched target for its critical role in selective autophagy. In this study, we aimed to identify FDA-approved drugs that bind to the LIR motif of p62 and inhibit its LIR function, which could be useful targets for modulating autophagy. To this, the homology model of the p62 protein was predicted using biological data, and docking analysis was performed using Molegro Virtual Docker and PyRx softwares. We further assessed the toxicity profile of the drugs using the ProTox-II server and performed dynamics simulations on the effective candidate drugs identified. The results revealed that the kanamycin, velpatasvir, verteporfin, and temoporfin significantly decreased the binding of LIR to the p62 protein. Finally, we experimentally confirmed that Kanamycin can inhibit autophagy-associated acidic vesicular formation in breast cancer MCF-7 and MDA-MB 231 cells. These repositioned drugs may represent novel autophagy modulators in clinical management, warranting further investigation.
Collapse
Affiliation(s)
- Narjes Asghari
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran
| | - Ali Kian Saei
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid 28040, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid 28040, Spain
| | - Zahra Nayeri
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box 14965/161, Tehran, Iran.
| |
Collapse
|
30
|
Kanmani S, Song XM, Kanmani P, Wu XJ, Xiao-Di-Tan, Liu J, Wang JP, Minshall RD, Hu G. Enhancement of Autophagy in Macrophages via the p120-Catenin-Mediated mTOR Signaling Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1666-1675. [PMID: 39423222 PMCID: PMC11610512 DOI: 10.4049/jimmunol.2400189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
Autophagy serves as a critical regulator of immune responses in sepsis. Macrophages are vital constituents of both innate and adaptive immunity. In this study, we delved into the intricate role of p120-catenin (p120) in orchestrating autophagy in macrophages in response to endotoxin stimulation. Depletion of p120 effectively suppressed LPS-induced autophagy in both J774A.1 macrophages and murine bone marrow-derived macrophages. LPS not only elevated the interaction between p120 and L chain 3 (LC3) I/II but also facilitated the association of p120 with mammalian target of rapamycin (mTOR). p120 depletion in macrophages by small interfering RNA reduced LPS-induced dissociation of mTOR and Unc-51-like kinase 1 (ULK1), leading to an increase in the phosphorylation of ULK1. p120 depletion also enhanced LPS-triggered macrophage apoptosis, as evidenced by increased levels of cleaved caspase 3, 7-aminoactinomycin D staining, and TUNEL assay. Notably, inhibiting autophagy reversed the decrease in apoptosis caused by LPS stimulation in macrophages overexpressing p120. Additionally, the ablation of p120 inhibited autophagy and accentuated apoptosis in alveolar macrophages in LPS-challenged mice. Collectively, our findings strongly suggest that p120 plays a pivotal role in fostering autophagy while concurrently hindering apoptosis in macrophages, achieved through modulation of the mTOR/ULK1 signaling pathway in sepsis. This underscores the potential of targeting macrophage p120 as an innovative therapeutic avenue for treating inflammatory disorders.
Collapse
Affiliation(s)
- Suganya Kanmani
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Xue-Min Song
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- The Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Wuchang, 125 Donghu Road, Hubei Province, China
| | - Paulraj Kanmani
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Xiao-Jing Wu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Wuchang, 238 Liberation Road, Hubei Province, China
| | - Xiao-Di-Tan
- Department Pediatrics, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Jing Liu
- Department of Surgery/Cancer Center, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Ji-Ping Wang
- Departments of Statistics and Data Science, Northwestern University, Evanston, Illinois, United States of America
| | - Richard D. Minshall
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, 60612, United States of America
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, 60612, United States of America
| |
Collapse
|
31
|
García Porta C, Mahfooz K, Komorowska J, Garcia-Rates S, Greenfield S. A Novel 14mer Peptide Inhibits Autophagic Flux via Selective Activation of the mTORC1 Signalling Pathway: Implications for Alzheimer's Disease. Int J Mol Sci 2024; 25:12837. [PMID: 39684549 DOI: 10.3390/ijms252312837] [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: 10/27/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
During development, a 14mer peptide, T14, modulates cell growth via the α-7 nicotinic acetylcholine receptor (α7 nAChR). However, this process could become excitotoxic in the context of the adult brain, leading to pathologies such as Alzheimer's disease (AD). Recent work shows that T14 acts selectively via the mammalian target of rapamycin complex 1 (mTORC1). This pathway is essential for normal development but is overactive in AD. The triggering of mTORC1 has also been associated with the suppression of autophagy, commonly observed in ageing and neurodegeneration. We therefore investigated the relationship between T14 and autophagic flux in tissue cultures, mouse brain slices, and human Alzheimer's disease hippocampus. Here, we demonstrate that T14 and p-mTOR s2448 expression significantly increases in AD human hippocampus, which was associated with the gradual decrease in the autophagosome number across Braak stages. During development, the reduction in T14 positively correlated with pTau (Ser202, Thr205) and two selective autophagy receptors: p62 and optineurin. In vitro studies also indicated that T14 increases p-mTOR s2448 expression, resulting in the aggregation of polyubiquinated substances. The effective blockade of T14 via its cyclic variant, NBP14, has been validated in vitro, in vivo, and ex vivo. In this study, NBP14 significantly attenuated p-mTOR s2448 expression and restored normal autophagic flux, as seen with rapamycin. We conclude that T14 acts at the α-7 receptor to selectively activate the mTORC1 pathway and consequently inhibit autophagic flux. Hence, this study describes a further step in the process by which T14 could drive neurodegeneration.
Collapse
Affiliation(s)
- Cloe García Porta
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Kashif Mahfooz
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Joanna Komorowska
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Sara Garcia-Rates
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Susan Greenfield
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| |
Collapse
|
32
|
Yu S, Lu X, Li C, Han Z, Li Y, Zhang X, Guo D. TFAM and Mitochondrial Protection in Diabetic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:4355-4365. [PMID: 39588133 PMCID: PMC11586499 DOI: 10.2147/dmso.s487815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic kidney disease (DKD) is a significant complication of diabetes and a major cause of end-stage renal disease. Affecting around 40% of diabetic patients, DKD poses substantial economic burdens due to its prevalence worldwide. The primary clinical features of DKD include the leakage of proteins into the urine, altered glomerular filtration, and an increased risk of cardiovascular diseases. Current treatments focus on managing hypertension and hyperglycemia to slow the progression of DKD. These include the use of SGLT2 inhibitors to control blood sugar and ACE inhibitors to reduce blood pressure. Despite these measures, current treatments do not cure DKD and fail to address its underlying causes. Emerging research highlights mitochondrial dysfunction as a pivotal factor in DKD progression. The kidneys' high energy requirements make them particularly susceptible to disturbances in mitochondrial function. In DKD, mitochondrial damage leads to reduced energy production and increased oxidative stress, exacerbating tissue damage. Mitochondrial DNA (mtDNA) damage is a key aspect of this dysfunction, with studies suggesting that changes in mtDNA copy number can serve as biomarkers for the progression of the disease. Efforts to target mitochondrial dysfunction are gaining traction as a potential therapeutic strategy. This includes promoting mitochondrial health through pharmacological and lifestyle interventions aimed at enhancing mitochondrial function and reducing oxidative stress. Such approaches could lead to more effective treatments that directly address the DKD.
Collapse
Affiliation(s)
- Siming Yu
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, People’s Republic of China
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xinxin Lu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chunsheng Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zehui Han
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yue Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xianlong Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Dandan Guo
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
33
|
Wei J, Wang X, Yu D, Tu Y, Yu Y. MicroRNA-mediated autophagy and drug resistance in cancer: mechanisms and therapeutic strategies. Discov Oncol 2024; 15:662. [PMID: 39549162 PMCID: PMC11569378 DOI: 10.1007/s12672-024-01525-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
This paper provides an exhaustive overview of the intricate interplay between microRNAs (miRNAs) and autophagy in the context of human cancers, underscoring the pivotal role these non-coding RNAs play in modulating autophagic pathways and their implications for cancer development, progression, and resistance to therapy. MiRNAs, as critical regulators of gene expression post-transcription, influence various biological processes, including autophagy, a catabolic mechanism essential for cellular homeostasis, stress response, and survival. The review meticulously delineates the mechanisms through which miRNAs impact autophagy by targeting specific genes and signaling pathways, thereby affecting cancer cell proliferation, metastasis, and response to chemotherapy. It highlights several miRNAs with dual roles, acting either as oncogenes or tumor suppressors based on the cellular context and the specific autophagic pathways they regulate. The paper further explores the therapeutic potential of targeting miRNA-autophagy axis, offering insights into novel strategies for cancer treatment through modulation of this axis. Emphasizing the complexity of the miRNA-autophagy relationship, the review calls for more in-depth studies to unravel the nuanced regulatory networks between miRNAs and autophagy in cancer, which could pave the way for the development of innovative therapeutic interventions and diagnostic tools.
Collapse
Affiliation(s)
- Jinxing Wei
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China
| | - Xianghui Wang
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China
| | - Duo Yu
- Department of Biopharmaceutics School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yanyang Tu
- Research Center, The Huizhou Central People's Hospital, Guangdong Medical University, No. 41 Eling North Road, Huizhou, Guangdong, China.
| | - Yaoyu Yu
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China.
| |
Collapse
|
34
|
Ke PY, Yeh CT. Functional Role of Hepatitis C Virus NS5A in the Regulation of Autophagy. Pathogens 2024; 13:980. [PMID: 39599533 PMCID: PMC11597459 DOI: 10.3390/pathogens13110980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Many types of RNA viruses, including the hepatitis C virus (HCV), activate autophagy in infected cells to promote viral growth and counteract the host defense response. Autophagy acts as a catabolic pathway in which unnecessary materials are removed via the lysosome, thus maintaining cellular homeostasis. The HCV non-structural 5A (NS5A) protein is a phosphoprotein required for viral RNA replication, virion assembly, and the determination of interferon (IFN) sensitivity. Recently, increasing evidence has shown that HCV NS5A can induce autophagy to promote mitochondrial turnover and the degradation of hepatocyte nuclear factor 1 alpha (HNF-1α) and diacylglycerol acyltransferase 1 (DGAT1). In this review, we summarize recent progress in understanding the detailed mechanism by which HCV NS5A triggers autophagy, and outline the physiological significance of the balance between host-virus interactions.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry and Molecular Biology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| |
Collapse
|
35
|
Ganguly U, Carroll T, Nehrke K, Johnson GVW. Mitochondrial Quality Control in Alzheimer's Disease: Insights from Caenorhabditis elegans Models. Antioxidants (Basel) 2024; 13:1343. [PMID: 39594485 PMCID: PMC11590956 DOI: 10.3390/antiox13111343] [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: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that is classically defined by the extracellular deposition of senile plaques rich in amyloid-beta (Aβ) protein and the intracellular accumulation of neurofibrillary tangles (NFTs) that are rich in aberrantly modified tau protein. In addition to aggregative and proteostatic abnormalities, neurons affected by AD also frequently possess dysfunctional mitochondria and disrupted mitochondrial maintenance, such as the inability to eliminate damaged mitochondria via mitophagy. Decades have been spent interrogating the etiopathogenesis of AD, and contributions from model organism research have aided in developing a more fundamental understanding of molecular dysfunction caused by Aβ and toxic tau aggregates. The soil nematode C. elegans is a genetic model organism that has been widely used for interrogating neurodegenerative mechanisms including AD. In this review, we discuss the advantages and limitations of the many C. elegans AD models, with a special focus and discussion on how mitochondrial quality control pathways (namely mitophagy) may contribute to AD development. We also summarize evidence on how targeting mitophagy has been therapeutically beneficial in AD. Lastly, we delineate possible mechanisms that can work alone or in concert to ultimately lead to mitophagy impairment in neurons and may contribute to AD etiopathology.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Trae Carroll
- Department of Pathology, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| |
Collapse
|
36
|
Anang V, Antonescu L, Nho R, Soni S, Mebratu YA. Targeting the Ubiquitin Proteasome System to Combat Influenza A Virus: Hijacking the Cleanup Crew. Rev Med Virol 2024; 34:e70005. [PMID: 39516190 DOI: 10.1002/rmv.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Influenza A virus (IAV) remains a significant global public health threat, causing substantial illness and economic burden. Despite existing antiviral drugs, the emergence of resistant strains necessitates alternative therapeutic strategies. This review explores the complex interplay between the ubiquitin proteasome system (UPS) and IAV pathogenesis. We discuss how IAV manipulates the UPS to promote its lifecycle, while also highlighting how host cells utilise the UPS to counteract viral infection. Recent research on deubiquitinases as potential regulators of IAV infection is also addressed. By elucidating the multifaceted role of the UPS in IAV pathogenesis, this review aims to identify potential targets for novel therapeutic interventions.
Collapse
Affiliation(s)
- Vandana Anang
- Pulmonary, Critical Care Medicine, and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Laura Antonescu
- Pulmonary, Critical Care Medicine, and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Richard Nho
- Pulmonary, Critical Care Medicine, and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Sourabh Soni
- Pulmonary, Critical Care Medicine, and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Yohannes A Mebratu
- Pulmonary, Critical Care Medicine, and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
37
|
Weigert A, Herhaus L. Immune modulation through secretory autophagy. J Cell Biochem 2024; 125:e30427. [PMID: 37260061 DOI: 10.1002/jcb.30427] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/06/2023] [Accepted: 05/13/2023] [Indexed: 06/02/2023]
Abstract
Autophagy is a central mechanism of cellular homeostasis through the degradation of a wide range of cellular constituents. However, recent evidence suggests that autophagy actively provides information to neighboring cells via a process called secretory autophagy. Secretory autophagy couples the autophagy machinery to the secretion of cellular content via extracellular vesicles (EVs). EVs carry a variety of cargo, that reflect the pathophysiological state of the originating cells and have the potential to change the functional profile of recipient cells, to modulate cell biology. The immune system has evolved to maintain local and systemic homeostasis. It is able to sense a wide array of molecules signaling disturbed homeostasis, including EVs and their content. In this review, we explore the emerging concept of secretory autophagy as a means to communicate cellular, and in total tissue pathophysiological states to the immune system to initiate the restoration of tissue homeostasis. Understanding how autophagy mediates the secretion of immunogenic factors may hold great potential for therapeutic intervention.
Collapse
Affiliation(s)
- Andreas Weigert
- Institute of Biochemistry I, Goethe University School of Medicine, Frankfurt am Main, Germany
| | - Lina Herhaus
- Institute of Biochemistry II, Goethe University School of Medicine, Frankfurt am Main, Germany
| |
Collapse
|
38
|
Wang C, Luo H. Crosstalk Between Innate Immunity and Autophagy in Viral Myocarditis Leading to Dilated Cardiomyopathy. Rev Med Virol 2024; 34:e2586. [PMID: 39349889 DOI: 10.1002/rmv.2586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 11/08/2024]
Abstract
Viral myocarditis, characterised by inflammation of the heart muscle, presents a significant challenge to global public health, particularly affecting younger individuals and often progressing to dilated cardiomyopathy (DCM), a leading cause of heart failure. Despite ongoing research efforts, viable treatments for this condition remain elusive. Recent studies have shed light on the complex interplay between the innate immune response and autophagy mechanisms, revealing their pivotal roles in the pathogenesis of viral myocarditis and subsequent DCM development. This review aims to delve into the recent advancements in understanding the molecular mechanisms and pathways that intersect innate immunity and autophagy in the context of viral myocarditis. Furthermore, it explores the potential therapeutic implications of these findings, offering insights into promising avenues for the management and treatment of this debilitating condition.
Collapse
Affiliation(s)
- Chen Wang
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
39
|
Sun J, Dai W, Zhao S, Liu J, Zhang J, Xu J, He P. Response to the CO 2 concentrating mechanisms and transcriptional time series analysis of Ulva prolifera under inorganic carbon limitation. HARMFUL ALGAE 2024; 139:102727. [PMID: 39567081 DOI: 10.1016/j.hal.2024.102727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 11/22/2024]
Abstract
Ulva prolifera is a dominant species in green tides and has been affecting marine ecosystem for many years. Due to the low availability of CO2 in the environment, U. prolifera utilizes the CO2 concentrating mechanisms (CCMs) to increase intracellular inorganic carbon concentration. However, the transcriptional response mechanism and temporal changes of U. prolifera CCMs based on transcriptomics have not been thoroughly described. Therefore, we induced U. prolifera CCMs in a low CO2 environment to explore the dynamic regulation of CCMs expression under inadequate inorganic carbon supply. The results showed that inorganic carbon limitation increased the inorganic carbon affinity of U. prolifera, upregulating CCMs. The first 24 h of inorganic carbon environmental changes were the most active period for U. prolifera's expression regulation. U. prolifera gradually achieved a new steady state by regulating metabolic processes such as nucleic acids, energy, and ethylene-activated signaling pathways. In the carbon fixation system of U. prolifera, there are characteristics of both biophysical and biochemical CCMs. After 24 h of inorganic carbon limitation, the biophysical CCMs becomes more effective under conditions of inorganic carbon depletion. This study aids in exploring the CCMs of U. prolifera and their evolution in response to environmental changes.
Collapse
Affiliation(s)
- Jingyi Sun
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai 201306, China
| | - Wei Dai
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai 201306, China
| | - Shuang Zhao
- Ocean College, Fujian Polytechnic Normal University, Fuzhou 350300, China
| | - Jinlin Liu
- State Key Laboratory of Marine Geology, Tongji University, Shanghai 200092, China; Project Management Office of China National Scientific Seafloor Observatory, Tongji University, Shanghai 200092, China
| | - Jianheng Zhang
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Juntian Xu
- Jiangsu Key Laboratory for Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China.
| | - Peimin He
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
40
|
Park SW, Park JH, Choi H, Jeon P, Lee SH, Shin WD, Kim HJ, Lee JA, Jang DJ. Differential roles of N- and C-terminal LIR motifs in the catalytic activity and membrane targeting of RavZ and ATG4B proteins. BMB Rep 2024; 57:497-502. [PMID: 39384175 PMCID: PMC11608851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/08/2024] [Accepted: 09/02/2024] [Indexed: 10/11/2024] Open
Abstract
Mammalian ATG8 proteins (mATG8s) are essential for selective autophagy because they recruit various proteins with LC3- interacting region (LIR) motifs to autophagic membranes. The RavZ protein, secreted by Legionella pneumophila, and mammalian ATG4B possess functional LIR motifs that participate in lipidated mATG8 deconjugation on autophagic membranes. RavZ comprises three functional LIR motifs at the N- and Cterminal sides of its catalytic domain (CAD). This study demonstrated that LIR motifs at the N-terminal side of the CAD of RavZ are involved in autophagic membrane targeting and substrate recognition, while LIR motif at the C-terminal side facilitate autophagic membrane targeting. Our results also revealed that the C-terminal LIR motif in human ATG4B is pivotal in delipidating LC3B-phosphatidylethanolamine (PE), but it plays a minor role in pro-LC3B priming in the cytosol. Therefore, introducing a functional LIR motif to the N-terminal of ATG4B does not affect LC3B-PE delipidation. This study clearly described the position-dependent roles of LIR motifs in RavZ and ATG4B in cellular contexts. [BMB Reports 2024; 57(11): 497-502].
Collapse
Affiliation(s)
- Sang-Won Park
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju 37224, Korea
| | - Ju-Hui Park
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Haneul Choi
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Pureum Jeon
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Seung-Hwan Lee
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Won-Dong Shin
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju 37224, Korea
| | - Hun-Joo Kim
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon 34054, Korea
| | - Deok-Jin Jang
- Department of Vector Entomology, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
- Research Institute of Invertebrate Vector, Kyungpook National University, Sangju 37224, Korea
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| |
Collapse
|
41
|
Lin L, Lin Y, Han Z, Wang K, Zhou S, Wang Z, Wang S, Chen H. Understanding the molecular regulatory mechanisms of autophagy in lung disease pathogenesis. Front Immunol 2024; 15:1460023. [PMID: 39544928 PMCID: PMC11560454 DOI: 10.3389/fimmu.2024.1460023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/07/2024] [Indexed: 11/17/2024] Open
Abstract
Lung disease development involves multiple cellular processes, including inflammation, cell death, and proliferation. Research increasingly indicates that autophagy and its regulatory proteins can influence inflammation, programmed cell death, cell proliferation, and innate immune responses. Autophagy plays a vital role in the maintenance of homeostasis and the adaptation of eukaryotic cells to stress by enabling the chelation, transport, and degradation of subcellular components, including proteins and organelles. This process is essential for sustaining cellular balance and ensuring the health of the mitochondrial population. Recent studies have begun to explore the connection between autophagy and the development of different lung diseases. This article reviews the latest findings on the molecular regulatory mechanisms of autophagy in lung diseases, with an emphasis on potential targeted therapies for autophagy.
Collapse
Affiliation(s)
- Lin Lin
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- School of Medicine, Southeast University, Nanjing, China
- Science Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Ke Wang
- Department of Science and Education, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Shuwei Zhou
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing, China
| | - Zhanzhan Wang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Siyu Wang
- Department of Preventive Medicine, Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Haoran Chen
- Science Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
42
|
Chen Y, Barylko B, Eichorst J, Mueller J, Albanesi J. Identification of the GABARAP binding determinant in PI4K2A. Biosci Rep 2024; 44:BSR20240200. [PMID: 39344512 PMCID: PMC11499380 DOI: 10.1042/bsr20240200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024] Open
Abstract
GABARAP is a member of the ATG8 family of ubiquitin-like autophagy related proteins. It was initially discovered as a facilitator of GABA-A receptor translocation to the plasma membrane and has since been shown to promote the intracellular transport of a variety of other proteins under non-autophagic conditions. We and others have shown that GABARAP interacts with the Type II phosphatidylinositol 4-kinase, PI4K2A, and that this interaction is important for autophagosome-lysosome fusion. Here, we identify a 7-amino acid segment within the PI4K2A catalytic domain that contains the GABARAP interaction motif (GIM). This segment resides in an exposed loop that is not conserved in the other mammalian Type II PI 4-kinase, PI4K2B, explaining the specificity of GABARAP binding to the PI4K2A isoform. Mutation of the PI4K2A GIM inhibits GABARAP binding and PI4K2A-mediated recruitment of cytosolic GABARAP to subcellular organelles. We further show that GABARAP binds to mono-phosphorylated phosphoinositides, PI3P, PI4P, and PI5P, raising the possibility that these lipids contribute to the binding energies that drive GABARAP-protein interactions on membranes.
Collapse
Affiliation(s)
- Yan Chen
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, U.S.A
| | - Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| | - John P. Eichorst
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, U.S.A
| | - Joachim D. Mueller
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, U.S.A
| | - Joseph P. Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| |
Collapse
|
43
|
He L, Kwon D, Trnka MJ, Liu Y, Yang J, Li K, Totah RA, Johnson EF, Burlingame AL, Correia MA. Liver CYP4A autophagic-lysosomal degradation (ALD): A major role for the autophagic receptor SQSTM1/p62 through an uncommon target interaction site. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618315. [PMID: 39464120 PMCID: PMC11507770 DOI: 10.1101/2024.10.14.618315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The hepatic P450 hemoproteins CYPs 4A are typical N-terminally anchored Type I endoplasmic reticulum (ER)-proteins, that are inducible by hypolipidemic drugs and other "peroxisome proliferators". They are engaged in the ω-/ω-1-oxidation of various fatty acids including arachidonic acid, prostaglandins and leukotrienes and in the biotransformation of some therapeutic drugs. Herein we report that of the mammalian liver CYPs 4A, human CYP4A11 and mouse Cyp4a12a are preferential targets of the ER-lysosome-associated degradation (ERLAD). Consequently, these proteins are stabilized both as 1%Triton X100-soluble and -insoluble species in mouse hepatocytes and HepG2-cells deficient in the autophagic initiation ATG5-gene. Although these proteins exhibit surface LC3-interacting regions (LIRs) that would target them directly to the autophagosome, they nevertheless interact intimately with the autophagic receptor SQSTM1/p62. Through structural deletion analyses and site-directed mutagenesis, we have identified the Cyp4A-interacting p62 subdomain to lie between residues 170 and 233, which include its Traf6-binding and LIM-binding subdomains. Mice carrying a liver-specific genetic deletion of p62 residues 69-251 (p62Mut) that includes the CYP4A-interacting subdomain also exhibit Cyp4a-protein stabilization both as Triton X100-soluble and -insoluble species. Consistently, p62Mut mouse liver microsomes exhibit enhanced ω- and ω-1-hydroxylation of arachidonic acid to its physiologically active metabolites 19- and 20-HETEs relative to the corresponding wild-type mouse liver microsomes. Collectively, our findings suggest that any disruption of CYP4A ERLAD results in functionally active P450 protein and consequent production of proinflammatory metabolites on one hand, and insoluble aggregates on the other, which may contribute to pathological aggregates i.e. Mallory-Denk bodies/inclusions, hallmarks of many liver diseases.
Collapse
|
44
|
Loers G, Bork U, Schachner M. Functional Relationships between L1CAM, LC3, ATG12, and Aβ. Int J Mol Sci 2024; 25:10829. [PMID: 39409157 PMCID: PMC11476435 DOI: 10.3390/ijms251910829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
Abnormal protein accumulations in the brain are linked to aging and the pathogenesis of dementia of various types, including Alzheimer's disease. These accumulations can be reduced by cell indigenous mechanisms. Among these is autophagy, whereby proteins are transferred to lysosomes for degradation. Autophagic dysfunction hampers the elimination of pathogenic protein aggregations that contribute to cell death. We had observed that the adhesion molecule L1 interacts with microtubule-associated protein 1 light-chain 3 (LC3), which is needed for autophagy substrate selection. L1 increases cell survival in an LC3-dependent manner via its extracellular LC3 interacting region (LIR). L1 also interacts with Aβ and reduces the Aβ plaque load in an AD model mouse. Based on these results, we investigated whether L1 could contribute to autophagy of aggregated Aβ and its clearance. We here show that L1 interacts with autophagy-related protein 12 (ATG12) via its LIR domain, whereas interaction with ubiquitin-binding protein p62/SQSTM1 does not depend on LIR. Aβ, bound to L1, is carried to the autophagosome leading to Aβ elimination. Showing that the mitophagy-related L1-70 fragment is ubiquitinated, we expect that the p62/SQSTM1 pathway also contributes to Aβ elimination. We propose that enhancing L1 functions may contribute to therapy in humans.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ute Bork
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| |
Collapse
|
45
|
Liang W, Hu L, Dai F, Shi Y, Yang L, Li C. Calreticulin from Apostichopus japonicus relieves endoplasmic reticulum stress induced by Vibrio splendidus through autophagy. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109798. [PMID: 39084275 DOI: 10.1016/j.fsi.2024.109798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
When organisms are exposed to external stimuli, misfolded proteins accumulate continuously, resulting in endoplasmic reticulum (ER) stress. Autophagy is of great significance for eliminating aggregated proteins and maintaining cellular homeostasis. However, the molecular mechanism of activating autophagy in response to ER stress in sea cucumber is remain unclear. In the current study, we demonstrated that the pathogen Vibrio splendidus can cause ER stress in Apostichopus japonicus coelomocytes and identified a Ca2+ binding partner calreticulin (designated as AjCRT), which increased with the occurrence of ER stress. The nucleotide sequence analysis showed that the open reading frame of AjCRT was 1242 bp and encoded a 413-amino-acid residue polyprotein with calreticulin domains. The spatial expression analysis revealed that AjCRT was ubiquitously expressed in all examined tissues with large magnitude in the coelomocytes and was minimally expressed in muscle. Furthermore, silencing AjCRT in vivo could significantly exacerbate ER stress induced by V. splendidus and resulted in the significant reduction of coelomocyte autophagy. These findings indicate a calreticulin-based mechanism that positively regulates autophagy in response to ER stress induced by pathogen infection. The results will provide a basis for understanding the way of host alleviating ER stress through autophagy, and pharmacological approaches may have potential for managing ER stress induced by pathogen and related cellular disorders.
Collapse
Affiliation(s)
- Weikang Liang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, 315211, PR China
| | - Lincheng Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, 315211, PR China
| | - Fa Dai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, 315211, PR China
| | - Yue Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, 315211, PR China
| | - Lei Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, 315211, PR China.
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
46
|
Renteln M. Toward Systemic Lipofuscin Removal. Rejuvenation Res 2024; 27:171-179. [PMID: 39041624 DOI: 10.1089/rej.2024.0034] [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: 07/24/2024] Open
Abstract
Lipofuscin is indigestible garbage that accumulates in the autophagic vesicles and cytosol of postmitotic cells with age. Drs. Brunk and Terman postulated that lipofuscin accumulation is the main or at least a major driving factor in aging. They even posited that the evolution of memory is the reason why we get lipofuscin at all, as stable synaptic connections must be maintained over time, meaning that the somas of neurons must also remain in the same locale. In other words, they cannot dilute out their garbage over time through cell division. Mechanistically, their position certainly makes sense given that rendering a large percentage of a postmitotic cell's lysosomes useless must almost certainly negatively affect that cell and the surrounding microenvironment. It may be the case that lipofuscin accumulation is the main issue with regard to current age-related disease. Degradation in situ may be an insurmountable task currently. However, a method of systemic lipofuscin removal is discussed herein.
Collapse
Affiliation(s)
- Michael Renteln
- Molecular Genetics and Biochemistry from USC, Los Angeles, California, USA
| |
Collapse
|
47
|
Li G, Zhang J, Zhao Z, Wang J, Li J, Xu W, Cui Z, Sun P, Yuan H, Wang T, Li K, Bai X, Ma X, Li P, Fu Y, Cao Y, Bao H, Li D, Liu Z, Zhu N, Tang L, Lu Z. RNF144B negatively regulates antiviral immunity by targeting MDA5 for autophagic degradation. EMBO Rep 2024; 25:4594-4624. [PMID: 39285245 PMCID: PMC11467429 DOI: 10.1038/s44319-024-00256-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 08/14/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
As a RIG-I-like receptor, MDA5 plays a critical role in antiviral innate immunity by acting as a cytoplasmic double-stranded RNA sensor capable of initiating type I interferon pathways. Here, we show that RNF144B specifically interacts with MDA5 and promotes K27/K33-linked polyubiquitination of MDA5 at lysine 23 and lysine 43, which promotes autophagic degradation of MDA5 by p62. Rnf144b deficiency greatly promotes IFN production and inhibits EMCV replication in vivo. Importantly, Rnf144b-/- mice has a significantly higher overall survival rate than wild-type mice upon EMCV infection. Collectively, our results identify RNF144B as a negative regulator of innate antiviral response by targeting CARDs of MDA5 and mediating autophagic degradation of MDA5.
Collapse
Affiliation(s)
- Guoxiu Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
| | - Zhixun Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Jian Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Jiaoyang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Weihong Xu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Zhanding Cui
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Pu Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Hong Yuan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Kun Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Xingwen Bai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Xueqing Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Pinghua Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Yuanfang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Yimei Cao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Huifang Bao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Dong Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Zaixin Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Ning Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Lijie Tang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Zengjun Lu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
| |
Collapse
|
48
|
Xing Z, Jiang X, Chen Y, Wang T, Li X, Wei X, Fan Q, Yang J, Wu H, Cheng J, Cai R. Glutamine deprivation in glioblastoma stem cells triggers autophagic SIRT3 degradation to epigenetically restrict CD133 expression and stemness. Apoptosis 2024; 29:1619-1631. [PMID: 39068621 DOI: 10.1007/s10495-024-02003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2024] [Indexed: 07/30/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor, and glioblastoma stem cells (GSCs) are the primary cause of GBM heterogeneity, invasiveness, and resistance to therapy. Sirtuin 3 (SIRT3) is mainly localized in the mitochondrial matrix and plays an important role in maintaining GSC stemness through cooperative interaction with the chaperone protein tumor necrosis factor receptor-associated protein 1 (TRAP1) to modulate mitochondrial respiration and oxidative stress. The present study aimed to further elucidate the specific mechanisms by which SIRT3 influences GSC stemness, including whether SIRT3 serves as an autophagy substrate and the mechanism of SIRT3 degradation. We first found that SIRT3 is enriched in CD133+ GSCs. Further experiments revealed that in addition to promoting mitochondrial respiration and reducing oxidative stress, SIRT3 maintains GSC stemness by epigenetically regulating CD133 expression via succinate. More importantly, we found that SIRT3 is degraded through the autophagy-lysosome pathway during GSC differentiation into GBM bulk tumor cells. GSCs are highly dependent on glutamine for survival, and in these cells, we found that glutamine deprivation triggers autophagic SIRT3 degradation to restrict CD133 expression, thereby disrupting the stemness of GSCs. Together our results reveal a novel mechanism by which SIRT3 regulates GSC stemness. We propose that glutamine restriction to trigger autophagic SIRT3 degradation offers a strategy to eliminate GSCs, which combined with other treatment methods may overcome GBM resistance to therapy as well as relapse.
Collapse
Affiliation(s)
- Zhengcao Xing
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianguo Jiang
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yalan Chen
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohe Li
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyun Wei
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuju Fan
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Yang
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongmei Wu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China.
| | - Jinke Cheng
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rong Cai
- Department of Biochemistry & Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
49
|
Deater M, Lloyd RE. TDRD3 functions as a selective autophagy receptor with dual roles in autophagy and modulation of stress granule stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614367. [PMID: 39345463 PMCID: PMC11430058 DOI: 10.1101/2024.09.22.614367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Tudor Domain Containing 3 (TDRD3) is a methylarginine-reader protein that functions as a scaffold in the nucleus facilitating transcription, however TDRD3 is also recruited to stress granules (SGs) during the Integrated Stress Response (ISR) although its function therein remains largely unknown. We previously showed that TDRD3 is a novel antiviral restriction factor that is cleaved by virus 2A protease, and plays complex modulatory roles in both interferon and inflammatory signaling during stress and enterovirus infections. Here we have found that TDRD3 contains structural motifs similar to known selective autophagy receptors such as p62/SQSTM1, sharing ubiquitin associated domains (UBA) and LC3 interacting regions (LIR) that anchor cargo destined for autophagosomes to activated LC3 protein coating autophagosome membranes. This is of interest since enteroviruses hijack autophagy machinery to facilitate formation of viral replication factories, virus assembly and egress from the infected cell. Here we explored possible roles of TDRD3 in autophagy, hypothesizing that TDRD3 may function as a specialized selective autophagy receptor. We found that KO of TDRD3 in HeLa cells significantly reduces starvation induced autophagy, while its reintroduction restores it in a dose-dependent manner. Autophagy receptors are degraded during autophagy and expression levels decrease during this time. We found that TDRD3 levels decrease to the same extent as the autophagy receptor p62/SQSTM1 during autophagy, indicating autophagy-targeted turnover in that role. Knockout of TDRD3 or G3BP1 did not make significant changes in overall cell localization of LC3B or p62/SQSTM1, but did result in greater concentration of Lamp2 phagosome marker for phagosomes and phagolysosomes. To test the potential roles of TDRD3 in autophagic processes, we created a series of deletion mutants of TDRD3 lacking either UBA domain or the various LIR motifs that are predicted to interact with LC3B. Microscopic examination of starved cells expressing these variants of TDRD3 showed ΔLIR-TDRD3 had defects in colocalization with LC3B or Lamp2. Further, super resolution microscopy revealed ring structures with TDRD3 interfacing with p62/SQSTM1. In examination of arsenite induced stress granules we found recruitment of TDRD3 variants disrupted normally tight SG condensation, altered the decay rate of SGs upon release from stress and the kinetics of SG formation. We found evidence that the LIR3 motif on TDRD3 is involved in TDRD3 interaction with LC3B in coIP experiments, colocalization studies, and that this motif plays a key role in TDRD3 recruitment to SGs and SG resolution. Overall, these data support a functional role of TDRD3 in selective autophagy in a mode similar to p62/SQSTM1, with specific roles in SG stability and turnover. Enterovirus cleavage of TDRD3 likely affects both antiviral and autophagic responses that the virus controls for replication.
Collapse
Affiliation(s)
- Matthew Deater
- Department of Molecular Virology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Richard E Lloyd
- Department of Molecular Virology and Immunology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
50
|
Li CM, Kang J, Baek J, Kim Y, Park H, Jung YK. Cytosolic FKBPL and ER-resident CKAP4 co-regulates ER-phagy and protein secretion. Nat Commun 2024; 15:7886. [PMID: 39251576 PMCID: PMC11383940 DOI: 10.1038/s41467-024-52188-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
Endoplasmic reticulum quality control is crucial for maintaining cellular homeostasis and adapting to stress conditions. Although several ER-phagy receptors have been identified, the collaboration between cytosolic and ER-resident factors in ER fragmentation and ER-phagy regulation remains unclear. Here, we perform a phenotype-based gain-of-function screen and identify a cytosolic protein, FKBPL, functioning as an ER-phagy regulator. Overexpression of FKBPL triggers ER fragmentation and ER-phagy. FKBPL has multiple protein binding domains, can self-associate and might act as a scaffold connecting CKAP4 and LC3/GABARAPs. CKAP4 serves as a bridge between FKBPL and ER-phagy cargo. ER-phagy-inducing conditions increase FKBPL-CKAP4 interaction followed by FKBPL oligomerization at the ER, leading to ER-phagy. In addition, FKBPL-CKAP4 deficiency leads to Golgi disassembly and lysosome impairment, and an increase in ER-derived secretory vesicles and enhances cytosolic protein secretion via microvesicle shedding. Taken together, FKBPL with the aid of CKAP4 induces ER fragmentation and ER-phagy, and FKBPL-CKAP4 deficiency facilitates protein secretion.
Collapse
Affiliation(s)
- Cathena Meiling Li
- School of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Jaemin Kang
- School of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Jongyeon Baek
- School of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Heemin Park
- School of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Korea.
- Interdisciplinary Program in Neuroscience, Seoul National University, Gwanak-gu, Seoul, Korea.
| |
Collapse
|