1
|
Millard E, Tooze SA. ATG9 not just an Autophagy Related Protein. J Mol Biol 2025:169288. [PMID: 40513646 DOI: 10.1016/j.jmb.2025.169288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 05/27/2025] [Accepted: 06/06/2025] [Indexed: 06/16/2025]
Abstract
Autophagy proteins coordinate the biogenesis of a phagophore, the formation and maturation of an autophagosome. Genetic mutations of these proteins can result in dysregulated autophagy, stalled autophagosome biogenesis, and lead to cell death. ATG9, the sole transmembrane ATG (autophagy related) protein governs the nucleation of the phagophore. At a molecular level ATG9 has been shown to be a lipid scramblase capable of redistributing lipids across the lipid bilayer. ATG9-positive vesicles can also deliver lipid-modifying enzymes to alter the lipid composition of membranes. Both functions are required for autophagy. However, ATG proteins, including ATG9, play key molecular roles in pathways unrelated to autophagy. ATG9 has been shown to function in multiple pathways at the Golgi, plasma membrane, and lysosomes. ATG9 can also play an important role in immune signalling. The trafficking of ATG9 in ATG9-positive vesicles is essential to many of these pathways. In this review we highlight the functions of ATG9 in autophagy and autophagy-unrelated pathways, here referred to as "non-canonical functions", and summarise the broader role of ATG9A in cell homeostasis.
Collapse
Affiliation(s)
- Emily Millard
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
2
|
Wang M, Bai F, Zhao Q, Shi J, Hao Y, Wu J. Quantitative Proteomics Revealed the Molecular Regulatory Network of Lysine and the Effects of Lysine Supplementation on Sunit Skeletal Muscle Satellite Cells. Animals (Basel) 2025; 15:1425. [PMID: 40427302 PMCID: PMC12108402 DOI: 10.3390/ani15101425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/30/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Stimulating skeletal muscle satellite cells (SMSCs) with amino acids improves their proliferation and differentiation, enhancing skeletal muscle mass, thereby increasing lean meat rate. This study explored lysine (Lys)'s effects on SMSCs and their protein profiles in Sunit sheep. SMSCs were successfully isolated, assessing their survival and proliferation after Lys stimulation at varying concentrations using the CCK-8 assay. Western blotting revealed Lys-induced changes in myogenic differentiation protein expression, while immunocytochemistry detected α-Actinin and Myostatin within the SMSCs. TMT proteomics identified differentially expressed proteins, which underwent functional and interaction analyses, with RT-qPCR validating the corresponding gene expression. This study revealed that 4 mmol/L of Lys significantly boosted SMSC proliferation. A 24 h stimulation with this concentration reduced Myostatin expression, and increased MYOD1 and α-Actinin levels in the SMSCs. A proteomic analysis identified 577 differentially expressed proteins, primarily associated with lipoblast differentiation and muscle development, as highlighted by the GO enrichment analysis. A pathway analysis further demonstrated these proteins' involvement in the autophagy-lysosome and NOD-like receptor signaling pathways. Lys enhances SMSC proliferation, differentiation, and adipogenesis in Sunit sheep, exhibiting antioxidant properties and supporting muscle stability and amino acid metabolism. It may also have anti-inflammatory, anti-pyroptotic, and proteolysis-inhibitory effects, offering insights into muscle growth mechanisms through amino acid supplementation in ruminants.
Collapse
Affiliation(s)
- Mingxu Wang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (J.S.); (Y.H.)
| | - Fan Bai
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot 010031, China; (F.B.); (Q.Z.)
| | - Qinan Zhao
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot 010031, China; (F.B.); (Q.Z.)
| | - Jianan Shi
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (J.S.); (Y.H.)
| | - Yutian Hao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (J.S.); (Y.H.)
| | - Jindi Wu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (J.S.); (Y.H.)
| |
Collapse
|
3
|
Attanasio S. Autophagy in cancer and protein conformational disorders. FEBS Lett 2025. [PMID: 40342093 DOI: 10.1002/1873-3468.70061] [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: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Autophagy is a catabolic process by which cells maintain cellular homeostasis through the degradation of dysfunctional cytoplasmic components, such as toxic misfolded proteins and damaged organelles, within the lysosome. It is a multistep process that is tightly regulated by nutrient, energy, and stress-sensing mechanisms. Autophagy plays a pivotal role in various biological processes, including protein and organelle quality control, defense against pathogen infections, cell metabolism, and immune surveillance. As a result, autophagy dysfunction is linked to a variety of pathological conditions. The role of autophagy in cancer is complex and dynamic. Depending on the context, autophagy can have both tumor-suppressive and pro-tumorigenic effects. In contrast, its role is more clearly defined in protein conformational disorders, where autophagy serves as a mechanism to reduce toxic protein aggregation, thereby improving cellular homeostasis. Because autophagy-based therapies hold promising potential for the treatment of cancer and protein conformational disorders, this review will highlight the latest findings and advancements in these areas.
Collapse
Affiliation(s)
- Sergio Attanasio
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
4
|
Zheng W, Pu M, Zeng S, Zhang H, Wang Q, Chen T, Zhou T, Chang C, Neculai D, Liu W. S-palmitoylation modulates ATG2-dependent non-vesicular lipid transport during starvation-induced autophagy. EMBO J 2025; 44:2596-2619. [PMID: 40128367 PMCID: PMC12048663 DOI: 10.1038/s44318-025-00410-7] [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: 09/26/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/26/2025] Open
Abstract
Lipid transfer proteins mediate the non-vesicular transport of lipids at membrane contact sites to regulate the lipid composition of organelle membranes. Despite significant recent advances in our understanding of the structural basis for lipid transfer, its functional regulation remains unclear. In this study, we report that S-palmitoylation modulates the cellular function of ATG2, a rod-like lipid transfer protein responsible for transporting phospholipids from the endoplasmic reticulum (ER) to phagophores during autophagosome formation. During starvation-induced autophagy, ATG2A undergoes depalmitoylation as the balance between ZDHHC11-mediated palmitoylation and APT1-mediated depalmitoylation. Inhibition of ATG2A depalmitoylation leads to impaired autophagosome formation and disrupted autophagic flux. Further, in cell and in vitro analyses demonstrate that S-palmitoylation at the C-terminus of ATG2A anchors the C-terminus to the ER. Depalmitoylation detaches the C-terminus from the ER membrane, enabling it to interact with phagophores and promoting their growth. These findings elucidate a S-palmitoylation-dependent regulatory mechanism of cellular ATG2, which may represent a broad regulatory strategy for lipid transport mediated by bridge-like transporters within cells.
Collapse
Affiliation(s)
- Wenhui Zheng
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Maomao Pu
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Sai Zeng
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Hongtao Zhang
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qian Wang
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Tao Chen
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Tianhua Zhou
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Chunmei Chang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| | - Dante Neculai
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
| | - Wei Liu
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
- Department of Ultrasound Medicine and State Key laboratory Implantation Device, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China.
| |
Collapse
|
5
|
Campisi D, Hawkins N, Bonjour K, Wollert T. The Role of WIPI2, ATG16L1 and ATG12-ATG5 in Selective and Nonselective Autophagy. J Mol Biol 2025:169138. [PMID: 40221132 DOI: 10.1016/j.jmb.2025.169138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/24/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Autophagy is a conserved cellular recycling pathway that delivers damaged or superfluous cytoplasmic material to lysosomes for degradation. In response to cytotoxic stress or starvation, autophagy can also sequester bulk cytoplasm and deliver it to lysosomes to regenerate building blocks. In macroautophagy, a membrane cisterna termed phagophore that encloses autophagic cargo is generated. The formation of the phagophore depends on a conserved machinery of autophagy related proteins. The phosphatidylinositol(3)-phosphate binding protein WIPI2 facilitates the transition from phagophore initiation to phagophore expansion by recruiting the ATG12-ATG5-ATG16L1 complex to phagophores. This complex functions as an E3-ligase to conjugate ubiquitin-like ATG8 proteins to phagophore membranes, which promotes tethering of cargo to phagophore membranes, phagophore expansion, maturation and the fusion of autophagosomes with lysosomes. ATG16L1 also has important functions independently of ATG12-ATG5 in autophagy and beyond. In this review, we will summarize the functions of WIPI2 and ATG16L1 in selective and nonselective autophagy.
Collapse
Affiliation(s)
- Daniele Campisi
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - N'Toia Hawkins
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Kennedy Bonjour
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Thomas Wollert
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France.
| |
Collapse
|
6
|
Hanna MG, Rodriguez Cruz HO, Fujise K, Wu Y, Xu CS, Pang S, Li Z, Monetti M, De Camilli P. BLTP3A is associated with membranes of the late endocytic pathway and is an effector of CASM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.28.615015. [PMID: 39386594 PMCID: PMC11463362 DOI: 10.1101/2024.09.28.615015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Recent studies have identified a family of rod-shaped proteins thought to mediate lipid transfer at intracellular membrane contacts by a bridge-like mechanism. We show one such protein, bridge-like lipid transfer protein 3A (BLTP3A)/UHRF1BP1 binds VAMP7 vesicles via its C-terminal region and anchors them to lysosomes via its chorein domain containing N-terminal region to Rab7. Upon lysosome damage, BLTP3A-positive vesicles rapidly (within minutes) dissociate from lysosomes. Lysosome damage is known to activate the CASM (Conjugation of ATG8 to Single Membranes) pathway leading to lipidation and recruitment to lysosomes of mammalian ATG8 (mATG8) proteins. We find that this process drives the reassociation of BLTP3A with damaged lysosomes via an interaction of its LIR motif with mATG8 which coincides with a dissociation from the vesicles. Our findings reveal that BLTP3A is an effector of CASM, potentially as part of a mechanism to help repair or minimize lysosome damage.
Collapse
Affiliation(s)
- Michael G. Hanna
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Hely O. Rodriguez Cruz
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Kenshiro Fujise
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Yumei Wu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - C. Shan Xu
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - Song Pang
- Yale University School of Medicine, New Haven, CT
| | - Zhuonging Li
- Proteomics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mara Monetti
- Proteomics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| |
Collapse
|
7
|
Ge Y, Cao Y, Li F, Wang J, Liu Y, Guo W, Liu J, Fu S. Growth, fusion and degradation of lipid droplets: advances in lipid droplet regulatory protein. Arch Physiol Biochem 2025; 131:109-118. [PMID: 39115279 DOI: 10.1080/13813455.2024.2388779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/02/2024] [Accepted: 07/28/2024] [Indexed: 03/28/2025]
Abstract
Context: An adequate supply of energy is essential for the proper functioning of all life activities in living organisms. As organelles that store neutral lipids, lipid droplets (LDs) are involved in the synthesis and metabolism of lipids in cells and are also an important source of energy supply. Methods and mechanisms: A comprehensive summary of the literature was first carried out to screen for relevant proteins affecting the morphological size of LDs. The size of milk fat globules (MFGs) is directly influenced by the morphological size of LDs, which also controls the energy storage capacity of LDs. In this review, we detail the progress of research into the role of some protein in regulating the morphological size of LDs. Conclusion: It has been discovered that the number of protein are involved in the control of LD growth and degradation, such as Rab18-mediated local synthesis of triacylglycerol (TAG), cell death-inducing DFF45-like effector family proteins (CIDEs)-mediated atypical fusion between LDs, Stomatin protein-mediated LD fusion and autophagy-related proteins (ATGs)-mediated autophagic degradation of LDs. However, more studies are needed in the future to enrich the network of mechanisms that regulate the morphological size of LDs.
Collapse
Affiliation(s)
- Yusong Ge
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yu Cao
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Feng Li
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Jiaxin Wang
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yuhao Liu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Wenjin Guo
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Juxiong Liu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shoupeng Fu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
8
|
Fuller DM, Wu Y, Schueder F, Rasool B, Nag S, Korfhage JL, Garcia-Milian R, Melnyk KD, Bewersdorf J, De Camilli P, Melia TJ. ATG2A engages Rab1a and ARFGAP1 positive membranes during autophagosome biogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645038. [PMID: 40196537 PMCID: PMC11974814 DOI: 10.1101/2025.03.24.645038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Autophagosomes form from seed membranes that expand through bulk-lipid transport via the bridge-like lipid transporter ATG2. The origins of the seed membranes and their relationship to the lipid transport machinery are poorly understood. Using proximity labeling and a variety of fluorescence microscopy techniques, we show that ATG2A localizes to extra-Golgi ARFGAP1 puncta during autophagosome biogenesis. ARFGAP1 itself is dispensable during macroautophagy, but among other proteins associating to these membranes, we find that Rab1 is essential. ATG2A co-immunoprecipitates strongly with Rab1a, and siRNA-mediated depletion of Rab1 blocks autophagy downstream of LC3B lipidation, similar to ATG2A depletion. Further, when either autophagosome formation or the early secretory pathway is perturbed, ARFGAP1 and Rab1a accumulate at ectopic locations with autophagic machinery. Our results suggest that ATG2A engages a Rab1a complex on select early secretory membranes at an early stage in autophagosome biogenesis.
Collapse
Affiliation(s)
- Devin M. Fuller
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, 20 MD
| | - Yumei Wu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, 20 MD
- Department of Neuroscience, Yale University School of Medicine, New Haven CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| | - Florian Schueder
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT, USA
| | - Burha Rasool
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Shanta Nag
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Justin L. Korfhage
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Rolando Garcia-Milian
- Bioinformatics Support Hub, Yale Medical Library, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510
| | - Katerina D. Melnyk
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Joerg Bewersdorf
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Physics, Yale University, New Haven, CT, USA
| | - Pietro De Camilli
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, 20 MD
- Department of Neuroscience, Yale University School of Medicine, New Haven CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| | - Thomas J. Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, 20 MD
| |
Collapse
|
9
|
Rose K, Herrmann E, Kakudji E, Lizarrondo J, Celebi AY, Wilfling F, Lewis SC, Hurley JH. In situ cryo-ET visualization of mitochondrial depolarization and mitophagic engulfment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645001. [PMID: 40196634 PMCID: PMC11974748 DOI: 10.1101/2025.03.24.645001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Defective mitochondrial quality control in response to loss of mitochondrial membrane polarization is implicated in Parkinson's disease by mutations in PINK1 and PRKN. Application of in situ cryo-electron tomography (cryo-ET) made it possible to visualize the consequences of mitochondrial depolarization at higher resolution than heretofore attainable. Parkin-expressing U2OS cells were treated with the depolarizing agents oligomycin and antimycin A (OA), subjected to cryo-FIB milling, and mitochondrial structure was characterized by in situ cryo-ET. Phagophores were visualized in association with mitochondrial fragments. Bridge-like lipid transporter (BLTP) densities potentially corresponding to ATG2A were seen connected to mitophagic phagophores. Mitochondria in OA-treated cells were fragmented and devoid of matrix calcium phosphate crystals. The intermembrane gap of cristae was narrowed and the intermembrane volume reduced, and some fragments were devoid of cristae. A subpopulation of ATP synthases re-localized from cristae to the inner boundary membrane (IBM) apposed to the outer membrane (OMM). The structure of the dome-shaped prohibitin complex, a dodecamer of PHB1-PHB2 dimers, was determined in situ by sub-tomogram averaging in untreated and treated cells and found to exist in open and closed conformations, with the closed conformation is enriched by OA treatment. These findings provide a set of native snapshots of the manifold nano-structural consequences of mitochondrial depolarization and provide a baseline for future in situ dissection of Parkin-dependent mitophagy.
Collapse
Affiliation(s)
- Kevin Rose
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Eric Herrmann
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Eve Kakudji
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Javier Lizarrondo
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - A Yasemin Celebi
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Florian Wilfling
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Samantha C Lewis
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - James H Hurley
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
10
|
Zheng Z, Ji C, Zhao H, Zhao YG. ATG2A acts as a tether to regulate autophagosome-lysosome fusion in neural cells. Autophagy 2025:1-12. [PMID: 40083067 DOI: 10.1080/15548627.2025.2479427] [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: 10/03/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025] Open
Abstract
The macroautophagy/autophagy proteins ATG2A and ATG2B transfer lipids for phagophore membrane growth. They also form stable complexes with WDR45 and WDR45B. Our previous study demonstrated that WDR45 and WDR45B mediate autophagosome-lysosome fusion in neural cells. Given the defective autophagosome formation in cells lacking both ATG2s, their role in later autophagy stages is hard to explore. Here, we report that in neuroblastoma-derived Neuro-2a (N2a) cells, knocking down (KD) Atg2a, but not Atg2b, results in significant accumulation of SQSTM1/p62 and MAP1LC3-II/LC3-II, indicating impaired autophagy. Atg2a deficiency does not affect autophagosome formation, but reduces colocalization of autophagosomal LC3 with late endosomal/lysosomal RFP-RAB7, suggesting impaired autophagosome-lysosome fusion. ATG2A interacts with the SNARE proteins STX17, SNAP29, and VAMP8, facilitating their assembly. Overexpression of ATG2A partially rescues the autophagosome-lysosome fusion defects in Wdr45- and Wdr45b-deficient cells. ATG2 and another tether protein, EPG5, function partially redundantly in mediating autophagosome-lysosome fusion. Thus, ATG2A plays a key role in neural autophagy by tethering autophagosomes with lysosomes for fusion.Abbreviations: AAV: adeno-associated virus; ATG2Ar: RNAi-resistant ATG2A; Baf: bafilomycin A1; co-IP: co-immunoprecipitation; CQ: chloroquine; DKD: double knockdown; DKO: double knockout; ER: endoplasmic reticulum; KD: knockdown; KO: knockout; MIL: membrane-impermeable Halo ligand; MPL: membrane-permeable Halo ligand; N2a: Neuro-2a; NC negative control; PG: phagophore; PtdIns3K: phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; TEM: Transmission electron microscopy; TM: transmembrane domain; WT: wild-type.
Collapse
Affiliation(s)
- Ze Zheng
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, P.R. China
| | - Cuicui Ji
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- Department of Biology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, P.R. China
| | - Hongyu Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Yan G Zhao
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, P.R. China
| |
Collapse
|
11
|
Morcos A, Jung Y, Fuller RN, Bertucci A, Nguyen A, Zhang Q, Emge T, Boyle KE, Wall NR, Vazquez M. Seco-Duocarmycin SA in Aggressive Glioblastoma Cell Lines. Int J Mol Sci 2025; 26:2766. [PMID: 40141405 PMCID: PMC11943345 DOI: 10.3390/ijms26062766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is among the most lethal primary brain tumors and is characterized by significant cellular heterogeneity and resistance to conventional therapies. This study investigates the efficacy of seco-duocarmycin SA (seco-DSA), a novel DNA alkylating agent. Initial investigations using a colony formation assay revealed that seco-DSA exhibits remarkable potential with IC50 values lower than its natural DSA counterpart. Cell viability assay indicated that LN18 cells showed a markedly greater sensitivity to DSA than T98G cells. Furthermore, seco-DSA achieved its full cytotoxic effect within 8 h of drug incubation in GBM cell lines. Although seco-DSA induced a concentration-dependent increase in apoptotic cell death, the extent of apoptosis did not fully account for the observed decrease in cell viability. Instead, seco-DSA treatment resulted in significant cell cycle arrest in S and G2/M phases. These findings suggest that seco-DSA's cytotoxicity in GBM cells is primarily due to its ability to disrupt cell cycle progression, though the precise mechanisms of action remain to be fully established, and further research is needed. Proteomic analysis of treated cells also indicates dysregulation of proteins involved in senescence, apoptosis, and DNA repair, alluding to seco-DSA-induced arrest as a major mechanism of GBM disruption. Data are available via ProteomeXchange with the dataset identifier "PXD061023". Our reports promote the future exploration of seco-DSA's therapeutic potential, representing a critical step toward developing a more targeted and effective treatment for GBM.
Collapse
Affiliation(s)
- Ann Morcos
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA; (A.M.); (Y.J.); (A.B.); (T.E.)
- Division of Biochemistry, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yeonkyu Jung
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA; (A.M.); (Y.J.); (A.B.); (T.E.)
- Division of Biochemistry, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Ryan N. Fuller
- Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA;
| | - Antonella Bertucci
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA; (A.M.); (Y.J.); (A.B.); (T.E.)
- Nuclear Response & Analysis, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Amy Nguyen
- Proteomics Core, Institute for Integrative Genome Biology, University of California, Riverside, CA 92521, USA; (A.N.); (Q.Z.)
| | - Quanqing Zhang
- Proteomics Core, Institute for Integrative Genome Biology, University of California, Riverside, CA 92521, USA; (A.N.); (Q.Z.)
| | - Tobias Emge
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA; (A.M.); (Y.J.); (A.B.); (T.E.)
- Division of Biochemistry, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | | | - Nathan R. Wall
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA; (A.M.); (Y.J.); (A.B.); (T.E.)
- Division of Biochemistry, Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Marcelo Vazquez
- Department of Radiation Medicine, James M. Slater, MD Proton Treatment & Research Center, Loma Linda University Health, Loma Linda, CA 92350, USA; (A.M.); (Y.J.); (A.B.); (T.E.)
- Radiobiology & Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| |
Collapse
|
12
|
Dziurdzik SK, Sridhar V, Eng H, Neuman SD, Yan J, Davey M, Taubert S, Bashirullah A, Conibear E. Hoi1 targets the yeast BLTP2 protein to ER-PM contact sites to regulate lipid homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637747. [PMID: 39990326 PMCID: PMC11844476 DOI: 10.1101/2025.02.11.637747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Membrane contact sites between organelles are important for maintaining cellular lipid homeostasis. Members of the recently identified family of bridge-like lipid transfer proteins (BLTPs) span opposing membranes at these contact sites to enable the rapid transfer of bulk lipids between organelles. While the VPS13 and ATG2 family members use organelle-specific adaptors for membrane targeting, the mechanisms that regulate other bridge-like transporters remain unknown. Here, we identify the conserved protein Ybl086c, which we name Hoi1 (Hob interactor 1), as an adaptor that targets the yeast BLTP2-like proteins Fmp27/Hob1 and Hob2 to ER-PM contact sites. Two separate Hoi1 domains interface with alpha-helical projections that decorate the central hydrophobic channel on Fmp27, and loss of these interactions disrupts cellular sterol homeostasis. The mutant phenotypes of BLTP2 and HOI1 orthologs indicate these proteins act in a shared pathway in worms and flies. Together, this suggests that Hoi1-mediated recruitment of BLTP2-like proteins represents an evolutionarily conserved mechanism for regulating lipid transport at membrane contact sites.
Collapse
Affiliation(s)
- Samantha K. Dziurdzik
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada V5Z 4H4
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada V6H 3N1
| | - Vaishnavi Sridhar
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada V5Z 4H4
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6H 3N1
| | - Hailey Eng
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada V5Z 4H4
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6H 3N1
| | - Sarah D. Neuman
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA
| | - Junran Yan
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada V5Z 4H4
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6H 3N1
| | - Michael Davey
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada V5Z 4H4
| | - Stefan Taubert
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada V5Z 4H4
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada V6H 3N1
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6H 3N1
| | - Arash Bashirullah
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA
| | - Elizabeth Conibear
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada V5Z 4H4
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada V6H 3N1
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada V6H 3N1
| |
Collapse
|
13
|
Vona R, Cittadini C, Ortona E, Matarrese P. Sex Disparity in Cancer: Role of Autophagy and Estrogen Receptors. Cells 2025; 14:273. [PMID: 39996745 PMCID: PMC11854201 DOI: 10.3390/cells14040273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Autophagy, a cellular process essential for maintaining homeostasis, plays a fundamental role in recycling damaged components and in adapting to stress. The dysregulation of autophagy is implicated in numerous human diseases, including cancer, where it exhibits a dual role as both a suppressor and a promoter, depending on the context and the stage of tumor development. The significant sex differences observed in autophagic processes are determined by biological factors, such as genetic makeup and sex hormones. Estrogens, through their interaction with specific receptors, modulate autophagy and influence tumor progression, therapy resistance, and the immune response to tumors. In females, the escape from X inactivation and estrogen signaling may be responsible for the advantages, in terms of lower incidence and longer survival, observed in oncology. Women often show better responses to traditional chemotherapy, while men respond better to immunotherapy. The action of sex hormones on the immune system could contribute to these differences. However, women experience more severe adverse reactions to anticancer drugs. The estrogen/autophagy crosstalk-involved in multiple aspects of the tumor, i.e., development, progression and the response to therapy-deserves an in-depth study, as it could highlight sex-specific mechanisms useful for designing innovative and gender-tailored treatments from the perspective of precision medicine.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| | | | | | - Paola Matarrese
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| |
Collapse
|
14
|
Yaeger JDW, Sengupta S, Walz AL, Morita M, Morgan TK, Vermeer PD, Francis KR. Cholesterol deficiency directs autophagy-dependent secretion of extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632510. [PMID: 39829772 PMCID: PMC11741461 DOI: 10.1101/2025.01.11.632510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Extracellular vesicle (EV) secretion is an important, though not fully understood, intercellular communication process. Lipid metabolism has been shown to regulate EV activity, though the impact of specific lipid classes is unclear. Through analysis of small EVs (sEVs), we observe aberrant increases in sEV release within genetic models of cholesterol biosynthesis disorders, where cellular cholesterol is diminished. Inhibition of cholesterol synthesis at multiple synthetic steps mimics genetic models in terms of cholesterol reduction and sEVs secreted. Further analyses of sEVs from cholesterol-depleted cells revealed structural deficits and altered surface marker expression, though these sEVs were also more easily internalized by recipient cells. Transmission electron microscopy of cells with impaired cholesterol biosynthesis demonstrated multivesicular and multilamellar structures potentially associated with autophagic defects. We further found autophagic vesicles being redirected toward late endosomes at the expense of autophagolysosomes. Through CRISPR-mediated inhibition of autophagosome formation, we mechanistically determined that release of sEVs after cholesterol depletion is autophagy dependent. We conclude that cholesterol imbalance initiates autophagosome-dependent secretion of sEVs, which may have pathological relevance in diseases of cholesterol disequilibrium.
Collapse
Affiliation(s)
- Jazmine D. W. Yaeger
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Sonali Sengupta
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Austin L. Walz
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Mayu Morita
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Terry K. Morgan
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
- Center for Developmental Health, Oregon Health and Science University, Portland, OR 97239, USA
| | - Paola D. Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Surgery, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Kevin R. Francis
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
15
|
Azizian M, Tamaddon GH, Ashrafi M, Chahardahcherik M, Gharechahi F. Impact of carboxymethyl dextran-asparaginase in NALM-6 cell apoptosis and autophagy. IRANIAN JOURNAL OF VETERINARY RESEARCH 2025; 25:344-352. [PMID: 40386102 PMCID: PMC12085209 DOI: 10.22099/ijvr.2024.49166.7208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Background Acute lymphoblastic leukemia (ALL) is a malignant disorder in both humans and animals. L-asparaginase (L-ASNase) has limitations as a chemotherapy agent due to adverse effects and low serum stability. In a previous study, L-ASNase was chemically modified with carboxymethyl dextran to enhance its properties. Aims This study aimed to validate the potential of these modifications using the NALM-6 cell line. Methods NALM-6 cells were cultured and treated with various concentrations, including 0 IU/ml as negative control, 0.5, 1, 1.5, and 2 IU/ml of modified L-ASNase and L-ASNase. The optimal concentration was determined at specific intervals, and viability and metabolic activity were assessed through Trypan blue and MTT tests. Flow cytometry, using Annexin V/PI staining, was employed to evaluate apoptosis. Real-time RT-PCR techniques were used to determine changes in the expression of the ATG2B and LC3-II genes (important genes in autophagy), with data analysis conducted using PRISM software. Results The modified L-ASNase reduced the viability of NALM-6 cells and induced higher levels of apoptosis (P=0.001). Interestingly, the modified enzyme had a lesser impact on autophagy, which is important for avoiding treatment resistance. Conclusion The modified L-ASNase showed enhanced effectiveness in reducing the viability of NALM-6 cells and induced higher levels of apoptosis. Interestingly, the modified enzyme had a lesser effect on autophagy, which is important as excessive autophagy can lead to treatment resistance. These findings suggest that the modified L-ASNase may have the potential to be a more effective chemotherapeutic agent for ALL treatments.
Collapse
Affiliation(s)
- M. Azizian
- Ph.D. Student in Biochemistry, Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - G. H. Tamaddon
- Department of Laboratory Sciences, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences and Health Services, Shiraz, Iran
| | - M. Ashrafi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - M. Chahardahcherik
- Ph.D. Student in Biochemistry, Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - F. Gharechahi
- MSc Student in Hematology, Department of Laboratory Sciences, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences and Health Services, Shiraz, Iran
| |
Collapse
|
16
|
Lv L, Yang C, Zhang X, Chen T, Luo M, Yu G, Chen Q. Autophagy-related protein PlATG2 regulates the vegetative growth, sporangial cleavage, autophagosome formation, and pathogenicity of peronophythora litchii. Virulence 2024; 15:2322183. [PMID: 38438325 PMCID: PMC10913709 DOI: 10.1080/21505594.2024.2322183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/18/2024] [Indexed: 03/06/2024] Open
Abstract
Autophagy is an intracellular degradation process that is important for the development and pathogenicity of phytopathogenic fungi and for the defence response of plants. However, the molecular mechanisms underlying autophagy in the pathogenicity of the plant pathogenic oomycete Peronophythora litchii, the causal agent of litchi downy blight, have not been well characterized. In this study, the autophagy-related protein ATG2 homolog, PlATG2, was identified and characterized using a CRISPR/Cas9-mediated gene replacement strategy in P. litchii. A monodansylcadaverine (MDC) staining assay indicated that deletion of PlATG2 abolished autophagosome formation. Infection assays demonstrated that ΔPlatg2 mutants showed significantly impaired pathogenicity in litchi leaves and fruits. Further studies have revealed that PlATG2 participates in radial growth and asexual/sexual development of P. litchii. Moreover, zoospore release and cytoplasmic cleavage of sporangia were considerably lower in the ΔPlatg2 mutants than in the wild-type strain by FM4-64 staining. Taken together, our results revealed that PlATG2 plays a pivotal role in vegetative growth, sporangia and oospore production, zoospore release, sporangial cleavage, and plant infection of P. litchii. This study advances our understanding of the pathogenicity mechanisms of the phytopathogenic oomycete P. litchii and is conducive to the development of effective control strategies.
Collapse
Affiliation(s)
- Lin Lv
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Chengdong Yang
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Xue Zhang
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Taixu Chen
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Manfei Luo
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Ge Yu
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Qinghe Chen
- Hainan Yazhou Bay Seed Laboratory, College of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Sanya, China
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| |
Collapse
|
17
|
Angara RK, Sadi A, Gilk SD. A novel bacterial effector protein mediates ER-LD membrane contacts to regulate host lipid droplets. EMBO Rep 2024; 25:5331-5351. [PMID: 39333627 PMCID: PMC11624262 DOI: 10.1038/s44319-024-00266-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024] Open
Abstract
Effective intracellular communication between cellular organelles occurs at dedicated membrane contact sites (MCSs). Tether proteins are responsible for the establishment of MCSs, enabling direct communication between organelles to ensure organelle function and host cell homeostasis. While recent research has identified tether proteins in several bacterial pathogens, their functions have predominantly been associated with mediating inter-organelle communication between the bacteria containing vacuole (BCV) and the host endoplasmic reticulum (ER). Here, we identify a novel bacterial effector protein, CbEPF1, which acts as a molecular tether beyond the confines of the BCV and facilitates interactions between host cell organelles. Coxiella burnetii, an obligate intracellular bacterial pathogen, encodes the FFAT motif-containing protein CbEPF1 which localizes to host lipid droplets (LDs). CbEPF1 establishes inter-organelle contact sites between host LDs and the ER through its interactions with VAP family proteins. Intriguingly, CbEPF1 modulates growth of host LDs in a FFAT motif-dependent manner. These findings highlight the potential for bacterial effector proteins to impact host cellular homeostasis by manipulating inter-organelle communication beyond conventional BCVs.
Collapse
Affiliation(s)
- Rajendra Kumar Angara
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Arif Sadi
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stacey D Gilk
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
18
|
He T, Ji C, Zhang W, Li X, Liu Y, Wang X, Zhang H, Wang J. The COPII coat protein SEC24D is required for autophagosome closure in mammals. FEBS Lett 2024; 598:2897-2909. [PMID: 39056365 DOI: 10.1002/1873-3468.14983] [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: 10/31/2023] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 07/28/2024]
Abstract
Macroautophagy involves the encapsulation of cellular components within double-membrane autophagosomes for subsequent degradation in vacuoles or lysosomes. Coat protein complex II (COPII) vesicles serve as a membrane source for autophagosome formation. However, the specific role of SEC24D, an isoform of the COPII coat protein SEC24, in the macroautophagy pathway remains unclear. In this study, we demonstrate that SEC24D is indispensable for macroautophagy and important for autophagosome closure. Depletion of SEC24D leads to the accumulation of unsealed isolation membranes. Furthermore, under conditions of starvation, SEC24D interacts with casein kinase1 delta (CK1δ), a member of the casein kinase 1 family, and autophagy-related 9A (ATG9A). Collectively, our findings unveil the indispensable role of SEC24D in starvation-induced autophagy in mammalian cells.
Collapse
Affiliation(s)
- Tianlong He
- College of Chemistry and Life Science, Beijing University of Technology, China
| | - Cuicui Ji
- College of Chemistry and Life Science, Beijing University of Technology, China
| | - Wenting Zhang
- College of Chemistry and Life Science, Beijing University of Technology, China
| | - Xianghua Li
- College of Chemistry and Life Science, Beijing University of Technology, China
| | - Yukun Liu
- College of Chemistry and Life Science, Beijing University of Technology, China
| | - Xiaoli Wang
- College of Chemistry and Life Science, Beijing University of Technology, China
| | - Haolin Zhang
- College of Chemistry and Life Science, Beijing University of Technology, China
| | - Juan Wang
- College of Chemistry and Life Science, Beijing University of Technology, China
| |
Collapse
|
19
|
Choi J, Jang H, Xuan Z, Park D. Emerging roles of ATG9/ATG9A in autophagy: implications for cell and neurobiology. Autophagy 2024; 20:2373-2387. [PMID: 39099167 PMCID: PMC11572220 DOI: 10.1080/15548627.2024.2384349] [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: 01/07/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024] Open
Abstract
Atg9, the only transmembrane protein among many autophagy-related proteins, was first identified in the year 2000 in yeast. Two homologs of Atg9, ATG9A and ATG9B, have been found in mammals. While ATG9B shows a tissue-specific expression pattern, such as in the placenta and pituitary gland, ATG9A is ubiquitously expressed. Additionally, ATG9A deficiency leads to severe defects not only at the molecular and cellular levels but also at the organismal level, suggesting key and fundamental roles for ATG9A. The subcellular localization of ATG9A on small vesicles and its functional relevance to autophagy have suggested a potential role for ATG9A in the lipid supply during autophagosome biogenesis. Nevertheless, the precise role of ATG9A in the autophagic process has remained a long-standing mystery, especially in neurons. Recent findings, however, including structural, proteomic, and biochemical analyses, have provided new insights into its function in the expansion of the phagophore membrane. In this review, we aim to understand various aspects of ATG9 (in invertebrates and plants)/ATG9A (in mammals), including its localization, trafficking, and other functions, in nonneuronal cells and neurons by comparing recent discoveries related to ATG9/ATG9A and proposing directions for future research.Abbreviation: AP-4: adaptor protein complex 4; ATG: autophagy related; cKO: conditional knockout; CLA-1: CLArinet (functional homolog of cytomatrix at the active zone proteins piccolo and fife); cryo-EM: cryogenic electron microscopy; ER: endoplasmic reticulum; KO: knockout; PAS: phagophore assembly site; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SV: synaptic vesicle; TGN: trans-Golgi network; ULK: unc-51 like autophagy activating kinase; WIPI2: WD repeat domain, phosphoinositide interacting 2.
Collapse
Affiliation(s)
- Jiyoung Choi
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon, South Korea
| | - Haeun Jang
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea
| | - Zhao Xuan
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Daehun Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, South Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon, South Korea
| |
Collapse
|
20
|
Norell PN, Campisi D, Mohan J, Wollert T. Biogenesis of omegasomes and autophagosomes in mammalian autophagy. Biochem Soc Trans 2024; 52:2145-2155. [PMID: 39392358 PMCID: PMC11555699 DOI: 10.1042/bst20240015] [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: 10/12/2024]
Abstract
Autophagy is a highly conserved catabolic pathway that maintains cellular homeostasis by promoting the degradation of damaged or superfluous cytoplasmic material. A hallmark of autophagy is the generation of membrane cisternae that sequester autophagic cargo. Expansion of these structures allows cargo to be engulfed in a highly selective and exclusive manner. Cytotoxic stress or starvation induces the formation of autophagosomes that sequester bulk cytoplasm instead of selected cargo. This rather nonselective pathway is essential for maintaining vital cellular functions during adverse conditions and is thus a major stress response pathway. Both selective and nonselective autophagy rely on the same molecular machinery. However, due to the different nature of cargo to be sequestered, the involved molecular mechanisms are fundamentally different. Although intense research over the past decades has advanced our understanding of autophagy, fundamental questions remain to be addressed. This review will focus on molecular principles and open questions regarding the formation of omegasomes and phagophores in nonselective mammalian autophagy.
Collapse
Affiliation(s)
- Puck N. Norell
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Daniele Campisi
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Jagan Mohan
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Thomas Wollert
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| |
Collapse
|
21
|
Qin Y, Zhou G, Jiao F, Cheng C, Meng C, Wang L, Wu S, Fan C, Li J, Zhou B, Chu Y, Jiao H. Brucella mediates autophagy, inflammation, and apoptosis to escape host killing. Front Cell Infect Microbiol 2024; 14:1408407. [PMID: 39507949 PMCID: PMC11537862 DOI: 10.3389/fcimb.2024.1408407] [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: 03/28/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024] Open
Abstract
Brucellosis is a serious zoonosis caused by Brucella spp. infection, which not only seriously jeopardizes the health of humans and mammals, but also causes huge economic losses to the livestock industry. Brucella is a Gram-negative intracellular bacterium that relies primarily on its virulence factors and a variety of evolved survival strategies to replicate and proliferate within cells. Currently, the mechanisms of autophagy, inflammation, and apoptosis in Brucella-infected hosts are not fully understood and require further research and discussion. This review focuses on the relationship between Brucella and autophagy, inflammation, and apoptosis to provide the scientific basis for revealing the pathogenesis of Brucella.
Collapse
Affiliation(s)
- Yaqiong Qin
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Gengxu Zhou
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Fengyuan Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chuan Cheng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chi Meng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Lingjie Wang
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Shengping Wu
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Cailiang Fan
- The College of Veterinary Medicine, Southwest University, Chongqing, China
- Animal Epidemic Prevention and Control Center of Rongchang, Chongqing, China
| | - Jixiang Li
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bo Zhou
- Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Yuefeng Chu
- 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, Gansu, China
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
22
|
Kumar A, Yadav S, Choudhary V. The evolving landscape of ER-LD contact sites. Front Cell Dev Biol 2024; 12:1483902. [PMID: 39421023 PMCID: PMC11484260 DOI: 10.3389/fcell.2024.1483902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Lipid droplets (LDs) are evolutionarily conserved dynamic organelles that play an important role in cellular physiology. Growing evidence suggests that LD biogenesis occurs at discrete endoplasmic reticulum (ER) subdomains demarcated by the lipodystrophy protein, Seipin, lack of which impairs adipogenesis. However, the mechanisms of how these domains are selected is not completely known. These ER sites undergo ordered assembly of proteins and lipids to initiate LD biogenesis and facilitate establishment of ER-LD contact sites, a prerequisite for proper growth and maturation of droplets. LDs retain both physical and functional association with the ER throughout their lifecycle to facilitate bi-directional communication, such as exchange of proteins and lipids between the two organelles at these ER-LD contact sites. In recent years several molecular tethers have been identified that bridge ER and LDs together including few proteins that are found exclusively at these ER-LD contact interface. Here, we discuss recent advances in understanding the role of factors that ensure functionality of ER-LD contact site machinery for LD homeostasis.
Collapse
Affiliation(s)
| | | | - Vineet Choudhary
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
23
|
Acharya S, Liao S, Jung WJ, Kang YS, Moghaddam VA, Feitosa MF, Wojczynski MK, Lin S, Anema JA, Schwander K, Connell JO, Province MA, Brent MR. A methodology for gene level omics-WAS integration identifies genes influencing traits associated with cardiovascular risks: the Long Life Family Study. Hum Genet 2024; 143:1241-1252. [PMID: 39276247 PMCID: PMC11485042 DOI: 10.1007/s00439-024-02701-1] [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: 03/07/2024] [Accepted: 08/15/2024] [Indexed: 09/16/2024]
Abstract
The Long Life Family Study (LLFS) enrolled 4953 participants in 539 pedigrees displaying exceptional longevity. To identify genetic mechanisms that affect cardiovascular risks in the LLFS population, we developed a multi-omics integration pipeline and applied it to 11 traits associated with cardiovascular risks. Using our pipeline, we aggregated gene-level statistics from rare-variant analysis, GWAS, and gene expression-trait association by Correlated Meta-Analysis (CMA). Across all traits, CMA identified 64 significant genes after Bonferroni correction (p ≤ 2.8 × 10-7), 29 of which replicated in the Framingham Heart Study (FHS) cohort. Notably, 20 of the 29 replicated genes do not have a previously known trait-associated variant in the GWAS Catalog within 50 kb. Thirteen modules in Protein-Protein Interaction (PPI) networks are significantly enriched in genes with low meta-analysis p-values for at least one trait, three of which are replicated in the FHS cohort. The functional annotation of genes in these modules showed a significant over-representation of trait-related biological processes including sterol transport, protein-lipid complex remodeling, and immune response regulation. Among major findings, our results suggest a role of triglyceride-associated and mast-cell functional genes FCER1A, MS4A2, GATA2, HDC, and HRH4 in atherosclerosis risks. Our findings also suggest that lower expression of ATG2A, a gene we found to be associated with BMI, may be both a cause and consequence of obesity. Finally, our results suggest that ENPP3 may play an intermediary role in triglyceride-induced inflammation. Our pipeline is freely available and implemented in the Nextflow workflow language, making it easily runnable on any compute platform ( https://nf-co.re/omicsgenetraitassociation ).
Collapse
Affiliation(s)
- Sandeep Acharya
- Division of Computational and Data Sciences, Washington University, St Louis, MO, USA
| | - Shu Liao
- Department of Computer Science and Engineering, Washington University, St Louis, MO, USA
| | - Wooseok J Jung
- Department of Computer Science and Engineering, Washington University, St Louis, MO, USA
| | - Yu S Kang
- Department of Computer Science and Engineering, Washington University, St Louis, MO, USA
| | - Vaha Akbary Moghaddam
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Shiow Lin
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Jason A Anema
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Karen Schwander
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Jeff O Connell
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Michael A Province
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Michael R Brent
- Department of Computer Science and Engineering, Washington University, St Louis, MO, USA.
| |
Collapse
|
24
|
Kang Y, Pogany J, Nagy PD. Proviral role of ATG2 autophagy related protein in tomato bushy stunt virus replication through bulk phospholipid transfer into the viral replication organelle. Mol Biol Cell 2024; 35:ar124. [PMID: 39110527 PMCID: PMC11481700 DOI: 10.1091/mbc.e24-05-0236] [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: 06/03/2024] [Accepted: 07/30/2024] [Indexed: 09/21/2024] Open
Abstract
Subversion of cellular membranes and membrane proliferation are used by positive-strand RNA viruses to build viral replication organelles (VROs) that support virus replication. The biogenesis of the membranous VROs requires major changes in lipid metabolism and lipid transfer in infected cells. In this work, we show that tomato bushy stunt virus (TBSV) hijacks Atg2 autophagy related protein with bulk lipid transfer activity into VROs via interaction with TBSV p33 replication protein. Deletion of Atg2 in yeast and knockdown of Atg2 in Nicotiana benthamiana resulted in decreased TBSV replication. We found that subversion of Atg2 by TBSV was important to enrich VRO membranes with phosphatidylethanolamine (PE), phosphatidylserine (PS) and PI(3)P phosphoinositide. Interestingly, inhibition of autophagy did not affect the efficient recruitment of Atg2 into VROs, and overexpression of Atg2 enhanced TBSV replication, indicating autophagy-independent subversion of Atg2 by TBSV. These findings suggest that the proviral function of Atg2 lipid transfer protein is in VRO membrane proliferation. In addition, we find that Atg2 interacting partner Atg9 with membrane lipid-scramblase activity is also coopted for tombusvirus replication. Altogether, the subversion of Atg2 bridge-type lipid transfer protein provides a new mechanism for tombusviruses to greatly expand VRO membranes to support robust viral replication.
Collapse
Affiliation(s)
- Yuanrong Kang
- Department of Plant Pathology, University of Kentucky, Plant Science Building, Lexington, KY 40546
| | - Judit Pogany
- Department of Plant Pathology, University of Kentucky, Plant Science Building, Lexington, KY 40546
| | - Peter D. Nagy
- Department of Plant Pathology, University of Kentucky, Plant Science Building, Lexington, KY 40546
| |
Collapse
|
25
|
Shen Y, Li T, Sun C, Cheng X, Chen Z, Wang G, Yang X. Atg7 autophagy-independent role on governing neural stem cell fate could be potentially applied for regenerative medicine. Cell Death Differ 2024; 31:1375-1388. [PMID: 38898232 PMCID: PMC11445561 DOI: 10.1038/s41418-024-01330-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
A literature review showed that Atg7 biological role was associated with the development and pathogenesis of nervous system, but very few reports focused on Atg7 role on neurogenesis at the region of spinal cord, so that we are committed to explore the subject. Atg7 expression in neural tube is incrementally increased during neurogenesis. Atg7 neural-specific knockout mice demonstrated the impaired motor function and imbalance of neuronal and glial cell differentiation during neurogenesis, which was similarly confirmed in primary neurosphere culture and reversely verified by Atg7 overexpression in unilateral neural tubes of gastrula chicken embryos. Furthermore, activating autophagy in neural stem cells (NSCs) of neurospheres did not rescue Atg7 deficiency-suppressed neuronal differentiation, but Atg7 overexpression on the basis of autophagy inhibition could reverse Atg7 deficiency-suppressed neuronal differentiation, which provides evidence for the existence of Atg7 role of autophagy-independent function. The underlying mechanism is that Atg7 deficiency directly caused the alteration of cell cycle length of NSCs, which is controlled by Atg7 through specifically binding Mdm2, thereby affecting neuronal differentiation during neurogenesis. Eventually, the effect of overexpressing Atg7-promoting neuronal differentiation was proved in spinal cord injury model as well. Taken together, this study revealed that Atg7 was involved in regulating neurogenesis by a non-autophagic signaling process, and this finding also shed light on the potential application in regenerative medicine.
Collapse
Affiliation(s)
- Yao Shen
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Tingting Li
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Chengyang Sun
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhi Chen
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, 510632, China.
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, School of Medicine, Jinan University, Guangzhou, 510220, China.
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, 510632, China.
- Clinical Research Center, Clifford Hospital, Guangzhou, 511496, China.
| |
Collapse
|
26
|
Makar AN, Boraman A, Mosen P, Simpson JE, Marques J, Michelberger T, Aitken S, Wheeler AP, Winter D, von Kriegsheim A, Gammoh N. The V-ATPase complex component RNAseK is required for lysosomal hydrolase delivery and autophagosome degradation. Nat Commun 2024; 15:7743. [PMID: 39231962 PMCID: PMC11374810 DOI: 10.1038/s41467-024-52049-3] [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: 05/02/2023] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
Autophagy is a finely orchestrated process required for the lysosomal degradation of cytosolic components. The final degradation step is essential for clearing autophagic cargo and recycling macromolecules. Using a CRISPR/Cas9-based screen, we identify RNAseK, a highly conserved transmembrane protein, as a regulator of autophagosome degradation. Analyses of RNAseK knockout cells reveal that, while autophagosome maturation is intact, cargo degradation is severely disrupted. Importantly, lysosomal protease activity and acidification remain intact in the absence of RNAseK suggesting a specificity to autolysosome degradation. Analyses of lysosome fractions show reduced levels of a subset of hydrolases in the absence of RNAseK. Of these, the knockdown of PLD3 leads to a defect in autophagosome clearance. Furthermore, the lysosomal fraction of RNAseK-depleted cells exhibits an accumulation of the ESCRT-III complex component, VPS4a, which is required for the lysosomal targeting of PLD3. Altogether, here we identify a lysosomal hydrolase delivery pathway required for efficient autolysosome degradation.
Collapse
Affiliation(s)
- Agata N Makar
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, UK
| | - Alina Boraman
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, UK
| | - Peter Mosen
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Joanne E Simpson
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, UK
| | - Jair Marques
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, UK
| | - Tim Michelberger
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, UK
| | - Stuart Aitken
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Crewe Road South, University of Edinburgh, Edinburgh, UK
| | - Ann P Wheeler
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Crewe Road South, University of Edinburgh, Edinburgh, UK
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alex von Kriegsheim
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, UK
| | - Noor Gammoh
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, UK.
| |
Collapse
|
27
|
Holzer E, Martens S, Tulli S. The Role of ATG9 Vesicles in Autophagosome Biogenesis. J Mol Biol 2024; 436:168489. [PMID: 38342428 DOI: 10.1016/j.jmb.2024.168489] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Autophagy mediates the degradation and recycling of cellular material in the lysosomal system. Dysfunctional autophagy is associated with a plethora of diseases including uncontrolled infections, cancer and neurodegeneration. In macroautophagy (hereafter autophagy) this material is encapsulated in double membrane vesicles, the autophagosomes, which form upon induction of autophagy. The precursors to autophagosomes, referred to as phagophores, first appear as small flattened membrane cisternae, which gradually enclose the cargo material as they grow. The assembly of phagophores during autophagy initiation has been a major subject of investigation over the past decades. A special focus has been ATG9, the only conserved transmembrane protein among the core machinery. The majority of ATG9 localizes to small Golgi-derived vesicles. Here we review the recent advances and breakthroughs regarding our understanding of how ATG9 and the vesicles it resides in serve to assemble the autophagy machinery and to establish membrane contact sites for autophagosome biogenesis. We also highlight open questions in the field that need to be addressed in the years to come.
Collapse
Affiliation(s)
- Elisabeth Holzer
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Vienna, Austria; University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Campus-Vienna-Biocenter 1, Vienna, Austria.
| | - Sascha Martens
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Vienna, Austria; University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria.
| | - Susanna Tulli
- Max Perutz Labs, Vienna BioCenter Campus (VBC), Vienna, Austria; University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria.
| |
Collapse
|
28
|
Zhu Y, Wang X, Liu R. Bioinformatics proved the existence of potential hub genes activating autophagy to participate in cartilage degeneration in osteonecrosis of the femoral head. J Mol Histol 2024; 55:539-554. [PMID: 38758521 DOI: 10.1007/s10735-024-10200-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/01/2024] [Indexed: 05/18/2024]
Abstract
The obvious degeneration of articular cartilage occurs in the late stage of osteonecrosis of the femoral head (ONFH), which aggravates the condition of ONFH. This study aimed to demonstrate aberrant activation of autophagy processes in ONFH chondrocytes through bioinformatics and to predict and identify relevant hub genes and pathways. Differentially expressed genes (DEGs) were identified using R software in the GSE74089 dataset from the GEO database. DEGs were crossed with the Human Autophagy Database (HADb) autophagy genes to screen out autophagy-related differential genes (AT-DEGs). GSEA, GSVA, GO, and KEGG pathway enrichment analyses of AT-DEGs were performed. The STRING database was used to analyze the protein-protein interaction (PPI) of the AT-DEGs network, and the MCODE and CytoHubba plugin in the Cytoscape software was used to analyze the key gene cluster module and screen the hub genes. The PPI network of hub genes was constructed using the GeneMANIA database, and functional enrichment and gene connectivity categories were analyzed. The expression levels of hub genes of related genes in the ONFH patients were verified in the dataset GSE123568, and the protein expression was verified by immunohistochemistry in tissues. The analysis of DEGs revealed abnormal autophagy in ONFH cartilage. AT-DEGs in ONFH have special enrichment in macroautophagy, autophagosome membrane, and phosphatidylinositol-3-phosphate binding. In the GSE123568 dataset, it was also found that ATG2B, ATG4B, and UVRAG were all significantly upregulated in ONFH patients. By immunohistochemistry, it was verified that ATG2B, ATG4B, and UVRAG were significantly overexpressed. These three genes regulate the occurrence and extension of autophagosomes through the PI3KC3C pathway. Finally, we determined that chondrocytes in ONFH undergo positive regulation of autophagy through the corresponding pathways involved in three genes: ATG2B, ATG4B, and UVRAG.
Collapse
Affiliation(s)
- Yingkang Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710004, China
| | - Xianxuan Wang
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ruiyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
29
|
Kurganovs NJ, Engedal N. To eat or not to eat: a critical review on the role of autophagy in prostate carcinogenesis and prostate cancer therapeutics. Front Pharmacol 2024; 15:1419806. [PMID: 38910881 PMCID: PMC11190189 DOI: 10.3389/fphar.2024.1419806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Around 1 in 7 men will be diagnosed with prostate cancer during their lifetime. Many strides have been made in the understanding and treatment of this malignancy over the years, however, despite this; treatment resistance and disease progression remain major clinical concerns. Recent evidence indicate that autophagy can affect cancer formation, progression, and therapeutic resistance. Autophagy is an evolutionarily conserved process that can remove unnecessary or dysfunctional components of the cell as a response to metabolic or environmental stress. Due to the emerging importance of autophagy in cancer, targeting autophagy should be considered as a potential option in disease management. In this review, along with exploring the advances made on understanding the role of autophagy in prostate carcinogenesis and therapeutics, we will critically consider the conflicting evidence observed in the literature and suggest how to obtain stronger experimental evidence, as the application of current findings in clinical practice is presently not viable.
Collapse
Affiliation(s)
- Natalie Jayne Kurganovs
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Nikolai Engedal
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
30
|
Nähse V, Stenmark H, Schink KO. Omegasomes control formation, expansion, and closure of autophagosomes. Bioessays 2024; 46:e2400038. [PMID: 38724256 DOI: 10.1002/bies.202400038] [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: 02/15/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/28/2024]
Abstract
Autophagy, an essential cellular process for maintaining cellular homeostasis and eliminating harmful cytoplasmic objects, involves the de novo formation of double-membraned autophagosomes that engulf and degrade cellular debris, protein aggregates, damaged organelles, and pathogens. Central to this process is the phagophore, which forms from donor membranes rich in lipids synthesized at various cellular sites, including the endoplasmic reticulum (ER), which has emerged as a primary source. The ER-associated omegasomes, characterized by their distinctive omega-shaped structure and accumulation of phosphatidylinositol 3-phosphate (PI3P), play a pivotal role in autophagosome formation. Omegasomes are thought to serve as platforms for phagophore assembly by recruiting essential proteins such as DFCP1/ZFYVE1 and facilitating lipid transfer to expand the phagophore. Despite the critical importance of phagophore biogenesis, many aspects remain poorly understood, particularly the complete range of proteins involved in omegasome dynamics, and the detailed mechanisms of lipid transfer and membrane contact site formation.
Collapse
Affiliation(s)
- Viola Nähse
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Harald Stenmark
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kay O Schink
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
31
|
Wei B, Fu Y, Li X, Chen F, Zhang Y, Chen H, Tong M, Li L, Pan Y, Zhang S, Chen S, Liu X, Zhong Q. ANKFY1 bridges ATG2A-mediated lipid transfer from endosomes to phagophores. Cell Discov 2024; 10:43. [PMID: 38622126 PMCID: PMC11018839 DOI: 10.1038/s41421-024-00659-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 02/02/2024] [Indexed: 04/17/2024] Open
Abstract
Macroautophagy is a process that cells engulf cytosolic materials by autophagosomes and deliver them to lysosomes for degradation. The biogenesis of autophagosomes requires ATG2 as a lipid transfer protein to transport lipids from existing membranes to phagophores. It is generally believed that endoplasmic reticulum is the main source for lipid supply of the forming autophagosomes; whether ATG2 can transfer lipids from other organelles to phagophores remains elusive. In this study, we identified a new ATG2A-binding protein, ANKFY1. Depletion of this endosome-localized protein led to the impaired autophagosome growth and the reduced autophagy flux, which largely phenocopied ATG2A/B depletion. A pool of ANKFY1 co-localized with ATG2A between endosomes and phagophores and depletion of UVRAG, ANKFY1 or ATG2A/B led to reduction of PI3P distribution on phagophores. Purified recombinant ANKFY1 bound to PI3P on membrane through its FYVE domain and enhanced ATG2A-mediated lipid transfer between PI3P-containing liposomes. Therefore, we propose that ANKFY1 recruits ATG2A to PI3P-enriched endosomes and promotes ATG2A-mediated lipid transfer from endosomes to phagophores. This finding implicates a new lipid source for ATG2A-mediated phagophore expansion, where endosomes donate PI3P and other lipids to phagophores via lipid transfer.
Collapse
Affiliation(s)
- Bin Wei
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhui Fu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuzhi Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqing Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanmo Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mindan Tong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linsen Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Pan
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shen Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - She Chen
- National Institute of Biological Sciences, Beijing, China
| | - Xiaoxia Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
32
|
Basei FL, e Silva IR, Dias PRF, Ferezin CC, Peres de Oliveira A, Issayama LK, Moura LAR, da Silva FR, Kobarg J. The Mitochondrial Connection: The Nek Kinases' New Functional Axis in Mitochondrial Homeostasis. Cells 2024; 13:473. [PMID: 38534317 PMCID: PMC10969439 DOI: 10.3390/cells13060473] [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: 01/24/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Mitochondria provide energy for all cellular processes, including reactions associated with cell cycle progression, DNA damage repair, and cilia formation. Moreover, mitochondria participate in cell fate decisions between death and survival. Nek family members have already been implicated in DNA damage response, cilia formation, cell death, and cell cycle control. Here, we discuss the role of several Nek family members, namely Nek1, Nek4, Nek5, Nek6, and Nek10, which are not exclusively dedicated to cell cycle-related functions, in controlling mitochondrial functions. Specifically, we review the function of these Neks in mitochondrial respiration and dynamics, mtDNA maintenance, stress response, and cell death. Finally, we discuss the interplay of other cell cycle kinases in mitochondrial function and vice versa. Nek1, Nek5, and Nek6 are connected to the stress response, including ROS control, mtDNA repair, autophagy, and apoptosis. Nek4, in turn, seems to be related to mitochondrial dynamics, while Nek10 is involved with mitochondrial metabolism. Here, we propose that the participation of Neks in mitochondrial roles is a new functional axis for the Nek family.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jörg Kobarg
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083-871, Brazil; (F.L.B.); (P.R.F.D.)
| |
Collapse
|
33
|
Acharya S, Liao S, Jung WJ, Kang YS, Moghaddam VA, Feitosa M, Wojczynski M, Lin S, Anema JA, Schwander K, Connell JO, Province M, Brent MR. Multi-omics Integration Identifies Genes Influencing Traits Associated with Cardiovascular Risks: The Long Life Family Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.04.24303657. [PMID: 38496585 PMCID: PMC10942516 DOI: 10.1101/2024.03.04.24303657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The Long Life Family Study (LLFS) enrolled 4,953 participants in 539 pedigrees displaying exceptional longevity. To identify genetic mechanisms that affect cardiovascular risks in the LLFS population, we developed a multi-omics integration pipeline and applied it to 11 traits associated with cardiovascular risks. Using our pipeline, we aggregated gene-level statistics from rare-variant analysis, GWAS, and gene expression-trait association by Correlated Meta-Analysis (CMA). Across all traits, CMA identified 64 significant genes after Bonferroni correction (p ≤ 2.8×10-7), 29 of which replicated in the Framingham Heart Study (FHS) cohort. Notably, 20 of the 29 replicated genes do not have a previously known trait-associated variant in the GWAS Catalog within 50 kb. Thirteen modules in Protein-Protein Interaction (PPI) networks are significantly enriched in genes with low meta-analysis p-values for at least one trait, three of which are replicated in the FHS cohort. The functional annotation of genes in these modules showed a significant over-representation of trait-related biological processes including sterol transport, protein-lipid complex remodeling, and immune response regulation. Among major findings, our results suggest a role of triglyceride-associated and mast-cell functional genes FCER1A, MS4A2, GATA2, HDC, and HRH4 in atherosclerosis risks. Our findings also suggest that lower expression of ATG2A, a gene we found to be associated with BMI, may be both a cause and consequence of obesity. Finally, our results suggest that ENPP3 may play an intermediary role in triglyceride-induced inflammation. Our pipeline is freely available and implemented in the Nextflow workflow language, making it easily runnable on any compute platform (https://nf-co.re/omicsgenetraitassociation).
Collapse
Affiliation(s)
- Sandeep Acharya
- Division of Computational and Data Sciences, Washington University, St Louis, MO
| | - Shu Liao
- Department of Computer Science and Engineering, Washington University, St Louis, MO
| | - Wooseok J Jung
- Department of Computer Science and Engineering, Washington University, St Louis, MO
| | - Yu S Kang
- Department of Computer Science and Engineering, Washington University, St Louis, MO
| | - Vaha A Moghaddam
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO
| | - Mary Feitosa
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO
| | - Mary Wojczynski
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO
| | - Shiow Lin
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO
| | - Jason A Anema
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO
| | - Karen Schwander
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO
| | - Jeff O Connell
- Department of Medicine, University of Maryland, Baltimore, MD
| | - Mike Province
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO
| | - Michael R Brent
- Department of Computer Science and Engineering, Washington University, St Louis, MO
| |
Collapse
|
34
|
Wang W, Su Y, Qi R, Li H, Jiang H, Li F, Li B, Sun H. Indoxacarb triggers autophagy and apoptosis through ROS accumulation mediated by oxidative phosphorylation in the midgut of Bombyx mori. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 200:105812. [PMID: 38582584 DOI: 10.1016/j.pestbp.2024.105812] [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: 01/04/2024] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 04/08/2024]
Abstract
Indoxacarb has been widely utilized in agricultural pest management, posing a significant ecological threat to Bombyx mori, a non-target economic insect. In the present study, short-term exposure to low concentration of indoxacarb significantly suppressed the oxidative phosphorylation pathway, and resulted in an accumulation of reactive oxygen species (ROS) in the midgut of B. mori. While, the ATP content exhibited a declining trend but there was no significant change. Moreover, indoxacarb also significantly altered the transcription levels of six autophagy-related genes, and the transcription levels of ATG2, ATG8 and ATG9 were significantly up-regulated by 2.56-, 1.90-, and 3.36-fold, respectively. The protein levels of ATG8-I and ATG8-II and MDC-stained frozen sections further suggested an increase in autophagy. Furthermore, the protein level and enzyme activity of CASP4 showed a significant increase in accordance with the transcription levels of apoptosis-related genes, indicating the activation of the apoptotic signaling pathway. Meanwhile, the induction of apoptosis signals in the midgut cells triggered by indoxacarb was confirmed through TUNEL staining. These findings suggest that indoxacarb can promote the accumulation of ROS by inhibiting the oxidative phosphorylation pathway, thereby inducing autophagy and apoptosis in the midgut cells of B. mori.
Collapse
Affiliation(s)
- Wanwan Wang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Yue Su
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Ruinan Qi
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Hao Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Hongrui Jiang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Fanchi Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China; Guangxi Collaborative Innovation Center of Modern Sericulture and Silk, School of Chemistry and Bioengineering, Hechi University, Yizhou, China; Sericulture Institute of Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bing Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China; Guangxi Collaborative Innovation Center of Modern Sericulture and Silk, School of Chemistry and Bioengineering, Hechi University, Yizhou, China; Sericulture Institute of Soochow University, Suzhou, Jiangsu 215123, PR China.
| | - Haina Sun
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, PR China; Guangxi Collaborative Innovation Center of Modern Sericulture and Silk, School of Chemistry and Bioengineering, Hechi University, Yizhou, China; Sericulture Institute of Soochow University, Suzhou, Jiangsu 215123, PR China.
| |
Collapse
|
35
|
Mathiowetz AJ, Olzmann JA. Lipid droplets and cellular lipid flux. Nat Cell Biol 2024; 26:331-345. [PMID: 38454048 PMCID: PMC11228001 DOI: 10.1038/s41556-024-01364-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 01/24/2024] [Indexed: 03/09/2024]
Abstract
Lipid droplets are dynamic organelles that store neutral lipids, serve the metabolic needs of cells, and sequester lipids to prevent lipotoxicity and membrane damage. Here we review the current understanding of the mechanisms of lipid droplet biogenesis and turnover, the transfer of lipids and metabolites at membrane contact sites, and the role of lipid droplets in regulating fatty acid flux in lipotoxicity and cell death.
Collapse
Affiliation(s)
- Alyssa J Mathiowetz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
36
|
Ren Q, Sun Q, Fu J. Dysfunction of autophagy in high-fat diet-induced non-alcoholic fatty liver disease. Autophagy 2024; 20:221-241. [PMID: 37700498 PMCID: PMC10813589 DOI: 10.1080/15548627.2023.2254191] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023] Open
Abstract
ABBREVIATIONS ACOX1: acyl-CoA oxidase 1; ADH5: alcohol dehydrogenase 5 (class III), chi polypeptide; ADIPOQ: adiponectin, C1Q and collagen domain containing; ATG: autophagy related; BECN1: beclin 1; CRTC2: CREB regulated transcription coactivator 2; ER: endoplasmic reticulum; F2RL1: F2R like trypsin receptor 1; FA: fatty acid; FOXO1: forkhead box O1; GLP1R: glucagon like peptide 1 receptor; GRK2: G protein-coupled receptor kinase 2; GTPase: guanosine triphosphatase; HFD: high-fat diet; HSCs: hepatic stellate cells; HTRA2: HtrA serine peptidase 2; IRGM: immunity related GTPase M; KD: knockdown; KDM6B: lysine demethylase 6B; KO: knockout; LAMP2: lysosomal associated membrane protein 2; LAP: LC3-associated phagocytosis; LDs: lipid droplets; Li KO: liver-specific knockout; LSECs: liver sinusoidal endothelial cells; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAP3K5: mitogen-activated protein kinase kinase kinase 5; MED1: mediator complex subunit 1; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; NFE2L2: NFE2 like bZIP transcription factor 2; NOS3: nitric oxide synthase 3; NR1H3: nuclear receptor subfamily 1 group H member 3; OA: oleic acid; OE: overexpression; OSBPL8: oxysterol binding protein like 8; PA: palmitic acid; RUBCNL: rubicon like autophagy enhancer; PLIN2: perilipin 2; PLIN3: perilipin 3; PPARA: peroxisome proliferator activated receptor alpha; PRKAA2/AMPK: protein kinase AMP-activated catalytic subunit alpha 2; RAB: member RAS oncogene family; RPTOR: regulatory associated protein of MTOR complex 1; SCD: stearoyl-CoA desaturase; SIRT1: sirtuin 1; SIRT3: sirtuin 3; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; SQSTM1/p62: sequestosome 1; SREBF1: sterol regulatory element binding transcription factor 1;SREBF2: sterol regulatory element binding transcription factor 2; STING1: stimulator of interferon response cGAMP interactor 1; STX17: syntaxin 17; TAGs: triacylglycerols; TFEB: transcription factor EB; TP53/p53: tumor protein p53; ULK1: unc-51 like autophagy activating kinase 1; VMP1: vacuole membrane protein 1.
Collapse
Affiliation(s)
- Qiannan Ren
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiming Sun
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junfen Fu
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
37
|
Wan N, Hong Z, Parson MAH, Korfhage JL, Burke JE, Melia TJ, Reinisch KM. Spartin-mediated lipid transfer facilitates lipid droplet turnover. Proc Natl Acad Sci U S A 2024; 121:e2314093121. [PMID: 38190532 PMCID: PMC10801920 DOI: 10.1073/pnas.2314093121] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024] Open
Abstract
Lipid droplets (LDs) are organelles critical for energy storage and membrane lipid homeostasis, whose number and size are carefully regulated in response to cellular conditions. The molecular mechanisms underlying lipid droplet biogenesis and degradation, however, are not well understood. The Troyer syndrome protein spartin (SPG20) supports LD delivery to autophagosomes for turnover via lipophagy. Here, we characterize spartin as a lipid transfer protein whose transfer ability is required for LD degradation. Spartin copurifies with phospholipids and neutral lipids from cells and transfers phospholipids in vitro via its senescence domain. A senescence domain truncation that impairs lipid transfer in vitro also impairs LD turnover in cells while not affecting spartin association with either LDs or autophagosomes, supporting that spartin's lipid transfer ability is physiologically relevant. Our data indicate a role for spartin-mediated lipid transfer in LD turnover.
Collapse
Affiliation(s)
- Neng Wan
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Zhouping Hong
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Matthew A. H. Parson
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BCV8W2Y2, Canada
| | - Justin L. Korfhage
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - John E. Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BCV8W2Y2, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Thomas J. Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Karin M. Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| |
Collapse
|
38
|
Liénard C, Pintart A, Bomont P. Neuronal Autophagy: Regulations and Implications in Health and Disease. Cells 2024; 13:103. [PMID: 38201307 PMCID: PMC10778363 DOI: 10.3390/cells13010103] [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: 10/26/2023] [Revised: 12/02/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Autophagy is a major degradative pathway that plays a key role in sustaining cell homeostasis, integrity, and physiological functions. Macroautophagy, which ensures the clearance of cytoplasmic components engulfed in a double-membrane autophagosome that fuses with lysosomes, is orchestrated by a complex cascade of events. Autophagy has a particularly strong impact on the nervous system, and mutations in core components cause numerous neurological diseases. We first review the regulation of autophagy, from autophagosome biogenesis to lysosomal degradation and associated neurodevelopmental/neurodegenerative disorders. We then describe how this process is specifically regulated in the axon and in the somatodendritic compartment and how it is altered in diseases. In particular, we present the neuronal specificities of autophagy, with the spatial control of autophagosome biogenesis, the close relationship of maturation with axonal transport, and the regulation by synaptic activity. Finally, we discuss the physiological functions of autophagy in the nervous system, during development and in adulthood.
Collapse
Affiliation(s)
- Caroline Liénard
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
- CHU Montpellier, University of Montpellier, 34295 Montpellier, France
| | - Alexandre Pintart
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
| | - Pascale Bomont
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
| |
Collapse
|
39
|
Angara RK, Sladek MF, Gilk SD. ER-LD Membrane Contact Sites: A Budding Area in the Pathogen Survival Strategy. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241304196. [PMID: 39697586 PMCID: PMC11653285 DOI: 10.1177/25152564241304196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024]
Abstract
The endoplasmic reticulum (ER) and lipid droplets (LDs) are essential organelles involved in lipid synthesis, storage, and transport. Physical membrane contacts between the ER and LDs facilitate lipid and protein exchange and thus play a critical role in regulating cellular lipid homeostasis. Recent research has revealed that ER-LD membrane contact sites are targeted by pathogens seeking to exploit host lipid metabolic processes. Both viruses and bacteria manipulate ER-LD membrane contact sites to enhance their replication and survival within the host. This review discusses the research advancements elucidating the mechanisms by which pathogens manipulate the ER-LD contacts through protein molecular mimicry and host cell protein manipulation, thereby hijacking host lipid metabolic processes to facilitate pathogenesis. Understanding the crosstalk between ER and LDs during infection provides deeper insight into host lipid regulation and uncovers potential therapeutic targets for treating infectious diseases.
Collapse
Affiliation(s)
- Rajendra Kumar Angara
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Margaret F. Sladek
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stacey D. Gilk
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
40
|
Danieli A, Vucak G, Baccarini M, Martens S. Sequestration of translation initiation factors in p62 condensates. Cell Rep 2023; 42:113583. [PMID: 38096057 DOI: 10.1016/j.celrep.2023.113583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 10/20/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Selective autophagy mediates the removal of harmful material from the cytoplasm. This cargo material is selected by cargo receptors, which orchestrate its sequestration within double-membrane autophagosomes and subsequent lysosomal degradation. The cargo receptor p62/SQSTM1 is present in cytoplasmic condensates, and a fraction of them are constantly delivered into lysosomes. However, the molecular composition of the p62 condensates is incompletely understood. To obtain insights into their composition, we develop a method to isolate these condensates and find that p62 condensates are enriched in components of the translation machinery. Furthermore, p62 interacts with translation initiation factors, and eukaryotic initiation factor 2α (eIF2α) and eIF4E are degraded by autophagy in a p62-dependent manner. Thus, p62-mediated autophagy may in part be linked to down-regulation of translation initiation. The p62 condensate isolation protocol developed here may facilitate the study of their contribution to cellular quality control and their roles in health and disease.
Collapse
Affiliation(s)
- Alberto Danieli
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria.
| | - Georg Vucak
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Manuela Baccarini
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Sascha Martens
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria.
| |
Collapse
|
41
|
Wan N, Hong Z, Parson MAH, Korfhage J, Burke JE, Melia TJ, Reinisch KM. Spartin-mediated lipid transfer facilitates lipid droplet turnover. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569220. [PMID: 38076959 PMCID: PMC10705495 DOI: 10.1101/2023.11.29.569220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Lipid droplets (LDs) are organelles critical for energy storage and membrane lipid homeostasis, whose number and size are carefully regulated in response to cellular conditions. The molecular mechanisms underlying lipid droplet biogenesis and degradation, however, are not well understood. The Troyer syndrome protein spartin (SPG20) supports LD delivery to autophagosomes for turnover via lipophagy. Here, we characterize spartin as a lipid transfer protein whose transfer ability is required for LD degradation. Spartin co-purifies with phospholipids and neutral lipids from cells and transfers phospholipids in vitro via its senescence domain. A senescence domain truncation that impairs lipid transfer in vitro also impairs LD turnover in cells while not affecting spartin association with either LDs or autophagosomes, supporting that spartin's lipid transfer ability is physiologically relevant. Our data indicate a role for spartin-mediated lipid transfer in LD turnover.
Collapse
Affiliation(s)
- Neng Wan
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Zhouping Hong
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Matthew A. H. Parson
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada V8W2Y2
| | - Justin Korfhage
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - John E. Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada V8W2Y2
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | - Thomas J. Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Karin M. Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
42
|
Huang Y, Zou S, Zhan P, Hao Z, Lu Q, Jing W, Li Y, Xu Y, Wang H. Dinotefuran induces oxidative stress and autophagy on Bombyx mori silk gland: Toxic effects and implications for nontarget organisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 336:122470. [PMID: 37657723 DOI: 10.1016/j.envpol.2023.122470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/03/2023]
Abstract
Dinotefuran, a third-generation neonicotinoid insecticide, is widely utilized in agriculture for pest control; however, its environmental consequences and risks to non-target organisms remain largely unknown. Bombyx mori is an economically important insect and a good toxic detector for environmental assessments. In this study, ultrastructure analysis showed that dinotefuran exposure caused an increase in autophagic vesicles in the silk gland. Dinotefuran exposure triggered elevated levels of oxidative stress in silk glands. Reactive oxygen species, oxidized glutathione disulfide, glutathione peroxidase, the activities of UDP glucuronosyl-transferase and carboxylesterase were induced in the middle silk gland, while malondialdehyde, reactive oxygen species, superoxide dismutase , oxidized glutathione disulfide were increased in the posterior silk gland. Global transcription patterns revealed the physiological responses were induced by dinotefuran. Dinotefuran exposure substantially induced the expression levels of many genes involved in the mTOR and PI3K - Akt signaling pathways in the middle silk gland, whereas many differentially expressed genes involved in fatty acid and pyrimidine metabolism were found in the posterior silk gland. Additionally, functional, ultrastructural, and transcriptomic analysis indicate that dinotefuran exposure induced an increase of autophagy in the silk gland. This study illuminates the toxicity effects of dinotefuran exposure on silkworms and provides new insights into the underlying molecular toxicity mechanisms of dinotefuran to nontarget organisms.
Collapse
Affiliation(s)
- Yuxin Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Shiyu Zou
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Pengfei Zhan
- Huzhou Academy of Agricultural Sciences, Huzhou, China
| | - Zhihua Hao
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qingyu Lu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wenhui Jing
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yinghui Li
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yusong Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Huabing Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
43
|
Scroggs SLP, Bird EJ, Molik DC, Nayduch D. Vesicular Stomatitis Virus Elicits Early Transcriptome Response in Culicoides sonorensis Cells. Viruses 2023; 15:2108. [PMID: 37896885 PMCID: PMC10612082 DOI: 10.3390/v15102108] [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: 09/19/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Viruses that are transmitted by arthropods, or arboviruses, have evolved to successfully navigate both the invertebrate and vertebrate hosts, including their immune systems. Biting midges transmit several arboviruses including vesicular stomatitis virus (VSV). To study the interaction between VSV and midges, we characterized the transcriptomic responses of VSV-infected and mock-infected Culicoides sonorensis cells at 1, 8, 24, and 96 h post inoculation (HPI). The transcriptomic response of VSV-infected cells at 1 HPI was significant, but by 8 HPI there were no detectable differences between the transcriptome profiles of VSV-infected and mock-infected cells. Several genes involved in immunity were upregulated (ATG2B and TRAF4) or downregulated (SMAD6 and TOLL7) in VSV-treated cells at 1 HPI. These results indicate that VSV infection in midge cells produces an early immune response that quickly wanes, giving insight into in vivo C. sonorensis VSV tolerance that may underlie their permissiveness as vectors for this virus.
Collapse
Affiliation(s)
- Stacey L. P. Scroggs
- Arthropod-Borne Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS 66502, USA; (D.C.M.); (D.N.)
| | - Edward J. Bird
- Department of Entomology, Kansas State University, Manhattan, KS 66502, USA;
| | - David C. Molik
- Arthropod-Borne Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS 66502, USA; (D.C.M.); (D.N.)
| | - Dana Nayduch
- Arthropod-Borne Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, KS 66502, USA; (D.C.M.); (D.N.)
| |
Collapse
|
44
|
Hanna M, Guillén-Samander A, De Camilli P. RBG Motif Bridge-Like Lipid Transport Proteins: Structure, Functions, and Open Questions. Annu Rev Cell Dev Biol 2023; 39:409-434. [PMID: 37406299 DOI: 10.1146/annurev-cellbio-120420-014634] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
The life of eukaryotic cells requires the transport of lipids between membranes, which are separated by the aqueous environment of the cytosol. Vesicle-mediated traffic along the secretory and endocytic pathways and lipid transfer proteins (LTPs) cooperate in this transport. Until recently, known LTPs were shown to carry one or a few lipids at a time and were thought to mediate transport by shuttle-like mechanisms. Over the last few years, a new family of LTPs has been discovered that is defined by a repeating β-groove (RBG) rod-like structure with a hydrophobic channel running along their entire length. This structure and the localization of these proteins at membrane contact sites suggest a bridge-like mechanism of lipid transport. Mutations in some of these proteins result in neurodegenerative and developmental disorders. Here we review the known properties and well-established or putative physiological roles of these proteins, and we highlight the many questions that remain open about their functions.
Collapse
Affiliation(s)
- Michael Hanna
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andrés Guillén-Samander
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut, USA
- Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
45
|
Tencer AH, Yu Y, Causse SZ, Campbell GR, Klein BJ, Xuan H, Cartier J, Miles MA, Gaurav N, Zadoroznyj A, Holt TA, Wen H, Hawkins CJ, Spector SA, Dubrez L, Shi X, Kutateladze TG. Molecular basis for nuclear accumulation and targeting of the inhibitor of apoptosis BIRC2. Nat Struct Mol Biol 2023; 30:1265-1274. [PMID: 37524969 PMCID: PMC10702411 DOI: 10.1038/s41594-023-01044-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/26/2023] [Indexed: 08/02/2023]
Abstract
The inhibitor of apoptosis protein BIRC2 regulates fundamental cell death and survival signaling pathways. Here we show that BIRC2 accumulates in the nucleus via binding of its second and third BIR domains, BIRC2BIR2 and BIRC2BIR3, to the histone H3 tail and report the structure of the BIRC2BIR3-H3 complex. RNA-seq analysis reveals that the genes involved in interferon and defense response signaling and cell-cycle regulation are most affected by depletion of BIRC2. Overexpression of BIRC2 delays DNA damage repair and recovery of the cell-cycle progression. We describe the structural mechanism for targeting of BIRC2BIR3 by a potent but biochemically uncharacterized small molecule inhibitor LCL161 and demonstrate that LCL161 disrupts the association of endogenous BIRC2 with H3 and stimulates cell death in cancer cells. We further show that LCL161 mediates degradation of BIRC2 in human immunodeficiency virus type 1-infected human CD4+ T cells. Our findings provide mechanistic insights into the nuclear accumulation of and blocking BIRC2.
Collapse
Affiliation(s)
- Adam H Tencer
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yucong Yu
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Sebastien Z Causse
- Institut National de la Santé et de la Recherche Médicale (Inserm), LNC UMR1231, Dijon, France and Université de Bourgogne Franche-Comté, Dijon, France
| | - Grant R Campbell
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Brianna J Klein
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hongwen Xuan
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jessy Cartier
- Institut National de la Santé et de la Recherche Médicale (Inserm), LNC UMR1231, Dijon, France and Université de Bourgogne Franche-Comté, Dijon, France
| | - Mark A Miles
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Nitika Gaurav
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Aymeric Zadoroznyj
- Institut National de la Santé et de la Recherche Médicale (Inserm), LNC UMR1231, Dijon, France and Université de Bourgogne Franche-Comté, Dijon, France
| | - Tina A Holt
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hong Wen
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Christine J Hawkins
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Stephen A Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Rady Children's Hospital, San Diego, CA, USA.
| | - Laurence Dubrez
- Institut National de la Santé et de la Recherche Médicale (Inserm), LNC UMR1231, Dijon, France and Université de Bourgogne Franche-Comté, Dijon, France.
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA.
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
46
|
Korfhage JL, Wan N, Elhan H, Kauffman L, Pineda M, Fuller DM, Thiam AR, Reinisch KM, Melia TJ. ATG2A-mediated bridge-like lipid transport regulates lipid droplet accumulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553257. [PMID: 37645754 PMCID: PMC10461963 DOI: 10.1101/2023.08.14.553257] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
ATG2 proteins facilitate bulk lipid transport between membranes. ATG2 is an essential autophagy protein, but ATG2 also localizes to lipid droplets (LDs), and genetic depletion of ATG2 increases LD numbers while impairing fatty acid transport from LDs to mitochondria. How ATG2 supports LD homeostasis and whether lipid transport regulates this homeostasis remains unknown. Here we demonstrate that ATG2 is preferentially recruited to phospholipid monolayers such as those surrounding LDs rather than to phospholipid bilayers. In vitro, ATG2 can drive phospholipid transport from artificial LDs with rates that correlate with the binding affinities, such that phospholipids are moved much more efficiently when one of the ATG2-interacting structures is an artificial LD. ATG2 is thought to exhibit 'bridge-like" lipid transport, with lipids flowing across the protein between membranes. We mutated key amino acids within the bridge to form a transport-dead ATG2 mutant (TD-ATG2A) which we show specifically blocks bridge-like, but not shuttle-like, lipid transport in vitro. TD-ATG2A still localizes to LDs, but is unable to rescue LD accumulation in ATG2 knockout cells. Thus, ATG2 has a natural affinity for, and an enhanced activity upon LD surfaces and uses bridge-like lipid transport to support LD dynamics in cells.
Collapse
Affiliation(s)
- Justin L. Korfhage
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| | - Neng Wan
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| | - Helin Elhan
- Laboratoire de Physique de l’École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, 75005 Paris, France
| | - Lisa Kauffman
- Laboratoire de Physique de l’École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, 75005 Paris, France
| | - Mia Pineda
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| | - Devin M. Fuller
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| | - Abdou Rachid Thiam
- Laboratoire de Physique de l’École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, 75005 Paris, France
| | - Karin M. Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| | - Thomas J. Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
47
|
Shimizu T, Tamura N, Nishimura T, Saito C, Yamamoto H, Mizushima N. Comprehensive analysis of autophagic functions of WIPI family proteins and their implications for the pathogenesis of β-propeller associated neurodegeneration. Hum Mol Genet 2023; 32:2623-2637. [PMID: 37364041 PMCID: PMC10407718 DOI: 10.1093/hmg/ddad096] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
β-propellers that bind polyphosphoinositides (PROPPINs) are an autophagy-related protein family conserved throughout eukaryotes. The PROPPIN family includes Atg18, Atg21 and Hsv2 in yeast and WD-repeat protein interacting with phosphoinositides (WIPI)1-4 in mammals. Mutations in the WIPI genes are associated with human neuronal diseases, including β-propeller associated neurodegeneration (BPAN) caused by mutations in WDR45 (encoding WIPI4). In contrast to yeast PROPPINs, the functions of mammalian WIPI1-WIPI4 have not been systematically investigated. Although the involvement of WIPI2 in autophagy has been clearly shown, the functions of WIPI1, WIPI3 and WIPI4 in autophagy remain poorly understood. In this study, we comprehensively analyzed the roles of WIPI proteins by using WIPI-knockout (single, double and quadruple knockout) HEK293T cells and recently developed HaloTag-based reporters, which enable us to monitor autophagic flux sensitively and quantitatively. We found that WIPI2 was nearly essential for autophagy. Autophagic flux was unaffected or only slightly reduced by single deletion of WIPI3 (encoded by WDR45B) or WIPI4 but was profoundly reduced by double deletion of WIPI3 and WIPI4. Furthermore, we revealed variable effects of BPAN-related missense mutations on the autophagic activity of WIPI4. BPAN is characterized by neurodevelopmental and neurodegenerative abnormalities, and we found a possible association between the magnitude of the defect of the autophagic activity of WIPI4 mutants and the severity of neurodevelopmental symptoms. However, some of the BPAN-related missense mutations, which produce neurodegenerative signs, showed almost normal autophagic activity, suggesting that non-autophagic functions of WIPI4 may be related to neurodegeneration in BPAN.
Collapse
Affiliation(s)
- Takahiro Shimizu
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Norito Tamura
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Taki Nishimura
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- PRESTO, Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Chieko Saito
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hayashi Yamamoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Molecular Oncology, Nippon Medical School, Institute for Advanced Medical Sciences, Tokyo 113-8602, Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
48
|
Omrane M, Ben M'Barek K, Santinho A, Nguyen N, Nag S, Melia TJ, Thiam AR. LC3B is lipidated to large lipid droplets during prolonged starvation for noncanonical autophagy. Dev Cell 2023; 58:1266-1281.e7. [PMID: 37315562 PMCID: PMC10686041 DOI: 10.1016/j.devcel.2023.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/23/2022] [Accepted: 05/17/2023] [Indexed: 06/16/2023]
Abstract
Lipid droplets (LDs) store lipids that can be utilized during times of scarcity via autophagic and lysosomal pathways, but how LDs and autophagosomes interact remained unclear. Here, we discovered that the E2 autophagic enzyme, ATG3, localizes to the surface of certain ultra-large LDs in differentiated murine 3T3-L1 adipocytes or Huh7 human liver cells undergoing prolonged starvation. Subsequently, ATG3 lipidates microtubule-associated protein 1 light-chain 3B (LC3B) to these LDs. In vitro, ATG3 could bind alone to purified and artificial LDs to mediate this lipidation reaction. We observed that LC3B-lipidated LDs were consistently in close proximity to collections of LC3B-membranes and were lacking Plin1. This phenotype is distinct from macrolipophagy, but it required autophagy because it disappeared following ATG5 or Beclin1 knockout. Our data suggest that extended starvation triggers a noncanonical autophagy mechanism, similar to LC3B-associated phagocytosis, in which the surface of large LDs serves as an LC3B lipidation platform for autophagic processes.
Collapse
Affiliation(s)
- Mohyeddine Omrane
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, 75005 Paris, France
| | - Kalthoum Ben M'Barek
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, 75005 Paris, France
| | - Alexandre Santinho
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, 75005 Paris, France
| | - Nathan Nguyen
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shanta Nag
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Thomas J Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Abdou Rachid Thiam
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, 75005 Paris, France.
| |
Collapse
|
49
|
Chen M, Menon MC, Wang W, Fu J, Yi Z, Sun Z, Liu J, Li Z, Mou L, Banu K, Lee SW, Dai Y, Anandakrishnan N, Azeloglu EU, Lee K, Zhang W, Das B, He JC, Wei C. HCK induces macrophage activation to promote renal inflammation and fibrosis via suppression of autophagy. Nat Commun 2023; 14:4297. [PMID: 37463911 PMCID: PMC10354075 DOI: 10.1038/s41467-023-40086-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
Renal inflammation and fibrosis are the common pathways leading to progressive chronic kidney disease (CKD). We previously identified hematopoietic cell kinase (HCK) as upregulated in human chronic allograft injury promoting kidney fibrosis; however, the cellular source and molecular mechanisms are unclear. Here, using immunostaining and single cell sequencing data, we show that HCK expression is highly enriched in pro-inflammatory macrophages in diseased kidneys. HCK-knockout (KO) or HCK-inhibitor decreases macrophage M1-like pro-inflammatory polarization, proliferation, and migration in RAW264.7 cells and bone marrow-derived macrophages (BMDM). We identify an interaction between HCK and ATG2A and CBL, two autophagy-related proteins, inhibiting autophagy flux in macrophages. In vivo, both global or myeloid cell specific HCK-KO attenuates renal inflammation and fibrosis with reduces macrophage numbers, pro-inflammatory polarization and migration into unilateral ureteral obstruction (UUO) kidneys and unilateral ischemia reperfusion injury (IRI) models. Finally, we developed a selective boron containing HCK inhibitor which can reduce macrophage pro-inflammatory activity, proliferation, and migration in vitro, and attenuate kidney fibrosis in the UUO mice. The current study elucidates mechanisms downstream of HCK regulating macrophage activation and polarization via autophagy in CKD and identifies that selective HCK inhibitors could be potentially developed as a new therapy for renal fibrosis.
Collapse
Affiliation(s)
- Man Chen
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Critical Care Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
- Department of Critical Care Medicine, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Madhav C Menon
- Division of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Wenlin Wang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeguo Sun
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Liu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzhe Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lingyun Mou
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Khadija Banu
- Division of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Sui-Wan Lee
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ying Dai
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bhaskar Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA.
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Renal Section, James J. Peters VAMC, Bronx, NY, USA.
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
50
|
Lu Q, Xu S, Hao Z, Li Y, Huang Y, Ying S, Jing W, Zou S, Xu Y, Wang H. Dinotefuran exposure induces autophagy and apoptosis through oxidative stress in Bombyx mori. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131997. [PMID: 37423129 DOI: 10.1016/j.jhazmat.2023.131997] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/18/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
As a third-generation neonicotinoid insecticide, dinotefuran is extensively used in agriculture, and its residue in the environment has potential effects on nontarget organisms. However, the toxic effects of dinotefuran exposure on nontarget organism remain largely unknown. This study explored the toxic effects of sublethal dose of dinotefuran on Bombyx mori. Dinotefuran upregulated reactive oxygen species (ROS) and malondialdehyde (MDA) levels in the midgut and fat body of B. mori. Transcriptional analysis revealed that the expression levels of many autophagy and apoptosis-associated genes were significantly altered after dinotefuran exposure, consistent with ultrastructural changes. Moreover, the expression levels of autophagy-related proteins (ATG8-PE and ATG6) and apoptosis-related proteins (BmDredd and BmICE) were increased, whereas the expression level of an autophagic key protein (sequestosome 1) was decreased in the dinotefuran-exposed group. These results indicate that dinotefuran exposure leads to oxidative stress, autophagy, and apoptosis in B. mori. In addition, its effect on the fat body was apparently greater than that on the midgut. In contrast, pretreatment with an autophagy inhibitor effectively downregulated the expression levels of ATG6 and BmDredd, but induced the expression of sequestosome 1, suggesting that dinotefuran-induced autophagy may promote apoptosis. This study reveals that ROS generation regulates the impact of dinotefuran on the crosstalk between autophagy and apoptosis, laying the foundation for studying cell death processes such as autophagy and apoptosis induced by pesticides. Furthermore, this study provides a comprehensive insight into the toxicity of dinotefuran on silkworm and contributes to the ecological risk assessment of dinotefuran in nontarget organisms.
Collapse
Affiliation(s)
- Qingyu Lu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shiliang Xu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhihua Hao
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinghui Li
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuxin Huang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shuye Ying
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenhui Jing
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shiyu Zou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yusong Xu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huabing Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|