1
|
Mao Y, Zhu P, Wang J, Fan C, Yu Z, Yao L, He W, Li X, Zhou F, Gan M, Wu X, Geng D. Protective effects of cannabinoid receptor 2 on annulus fibrosus degeneration by upregulating autophagy via AKT-mTOR-p70S6K signal pathway. Biochem Pharmacol 2025; 232:116734. [PMID: 39710272 DOI: 10.1016/j.bcp.2024.116734] [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: 08/19/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
As an important pathological process, annulus fibrosus (AF) degeneration contributes greatly to intervertebral disc degeneration (IVDD). Moreover, extracellular matrix (ECM) degradation and AF cell (AFC) autophagy are of utmost importance. The involvement of cannabinoid receptor type 2 (CB2) in the pathological mechanisms underlying different diseases has been demonstrated dueto its capacity toregulateautophagy. The objective of this study was to explore the impact of CB2-induced autophagy on AF degeneration and its underlying mechanism. First, the expression of CB2 in human degenerative AF tissues decreased with increasing degeneration degree, whereas its expression in rat AFCs increased in a concentration- and time-dependent manner following H2O2 intervention. Activation of CB2 increased collagen Ⅰ and Ⅱ expression while decreasing MMP3 and MMP13 expression. In addition, p62 expression decreased, whereas beclin-1 and LC3-Ⅱ/LC3-Ⅰ expression increased after JWH133 intervention. After CB2 activation, the addition of 3-MA impeded the synthesis of collagen Ⅰ and Ⅱ while preserving the elevated levels of MMP3 and MMP13. The activation of CB2 greatly suppressed the protein levels of the AKT/mTOR/p70S6K signaling pathway. In vivo, the JWH133 group exhibited elevated disk height index (DHI) and MRI signals, along with a comparatively intact structure of the intervertebral disc in contrast to the vehicle group. In general, CB2 activation could modulate apoptosis and autophagy in rat AFCs, thereby mitigating the advancement of IVDD. Moreover, the AKT/mTOR/p70S6K signaling pathway plays a role in the development of AF degeneration through the regulation of autophagy. The findings suggest that CB2 is a potentially effective therapeutic target for IVDD.
Collapse
Affiliation(s)
- Yubo Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi 214000, Jiangsu, China
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Jiale Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Chunyang Fan
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Zilin Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Lingye Yao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Wei He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Xinyun Li
- Department of Orthopedics, Medical School of Nantong University Clinical Medicine, Nantong 226000, Jiangsu, China
| | - Feng Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| | - Minfeng Gan
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| | - Xiexing Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
2
|
Leytens A, Benítez-Fernández R, Jiménez-García C, Roubaty C, Stumpe M, Boya P, Dengjel J. Targeted proteomics addresses selectivity and complexity of protein degradation by autophagy. Autophagy 2025; 21:460-475. [PMID: 39245437 PMCID: PMC11759517 DOI: 10.1080/15548627.2024.2396792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Macroautophagy/autophagy is a constitutively active catabolic lysosomal degradation pathway, often found dysregulated in human diseases. It is often considered to act in a cytoprotective manner and is commonly upregulated in cells undergoing stress. Its initiation is regulated at the protein level and does not require de novo protein synthesis. Historically, autophagy has been regarded as nonselective; however, it is now clear that different stimuli can lead to the selective degradation of cellular components via selective autophagy receptors (SARs). Due to its selective nature and the existence of multiple degradation pathways potentially acting in concert, monitoring of autophagy flux, i.e. selective autophagy-dependent protein degradation, should address this complexity. Here, we introduce a targeted proteomics approach monitoring abundance changes of 37 autophagy-related proteins covering process-relevant proteins such as the initiation complex and the Atg8-family protein lipidation machinery, as well as most known SARs. We show that proteins involved in autophagosome biogenesis are upregulated and spared from degradation under autophagy-inducing conditions in contrast to SARs, in a cell-line dependent manner. Classical bulk stimuli such as nutrient starvation mainly induce degradation of ubiquitin-dependent soluble SARs and not of ubiquitin-independent, membrane-bound SARs. In contrast, treatment with the iron chelator deferiprone leads to the degradation of ubiquitin-dependent and -independent SARs linked to mitophagy and reticulophagy/ER-phagy. Our approach is automatable and supports large-scale screening assays paving the way to (pre)clinical applications and monitoring of specific autophagy flux.Abbreviation: AMBRA1: autophagy and beclin 1 regulator 1; ATG: autophagy related; BafA1: bafilomycin A1; BNIP1: BCL2 interacting protein 1; BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3-like; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CCPG1: cell cycle progression 1; CV: coefficients of variations; CCCP: carbonyl cyanide m-chlorophenyl hydrazone; DFP: deferiprone; ER: endoplasmic reticulum; FKBP8: FKBP prolyl isomerase 8; GABARAPL: GABA type A receptor associated protein like; LC: liquid chromatography; LOD: limit of detection; LOQ: limit of quantification; MAP1LC3: microtubule associated protein 1 light chain 3; MS: mass spectrometry; NCOA4: nuclear receptor coactivator 4; NBR1: NBR1 autophagy cargo receptor; NUFIP1: nuclear FMR1 interacting protein 1; OPTN: optineurin; PHB2: prohibitin 2; PNPLA2/ATGL: patatin like phospholipase domain containing 2; POI: protein of interest; PTM: posttranslational modification; PRM: parallel reaction monitoring; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RETREG1/FAM134B: reticulophagy regulator 1; RPS6KB1: ribosomal protein S6 kinase B1; RTN3: reticulon 3; SARs: selective autophagy receptors; SQSTM1/p62: sequestosome 1; STBD1: starch binding domain 1; TAX1BP1: Tax1 binding protein 1; TFEB: transcription factor EB; TNIP1: TNFAIP3 interacting protein 1; TOLLIP: toll interacting protein; ULK1: unc-51 like autophagy activating kinase 1; WBP2: WW domain binding protein 2; WDFY3/Alfy: WD repeat and FYVE domain containing 3; WIPI2: WD repeat domain, phosphoinositide interacting 2.
Collapse
Affiliation(s)
- Alexandre Leytens
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Rocío Benítez-Fernández
- Department of Neuroscience and Movement Science, University of Fribourg, Fribourg, Switzerland
| | - Carlos Jiménez-García
- Department of Neuroscience and Movement Science, University of Fribourg, Fribourg, Switzerland
| | - Carole Roubaty
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Michael Stumpe
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Patricia Boya
- Department of Neuroscience and Movement Science, University of Fribourg, Fribourg, Switzerland
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
3
|
Wen Y, Wang M, Liu X, Yin X, Gong S, Yin J. Deletion of FgAtg27 decreases the pathogenicity of Fusarium graminearum through influence autophagic process. Int J Biol Macromol 2025; 297:139818. [PMID: 39814284 DOI: 10.1016/j.ijbiomac.2025.139818] [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: 10/20/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Autophagy is a conserved and unique degradation system in eukaryotic cells, which plays crucial roles in the growth, development and pathogenesis of Fungi. Despite that, it is poorly understood in Fusarium graminearum currently. Here, we identified an autophagy gene FgAtg27 from F. graminearum, and investigated its possible roles in regulating morphogenesis and pathogenicity. Results showed that FgAtg27 is homologous to Saccharomyces cerevisiae Atg27 and with an active signal peptide at N-terminal. Then, the ΔFgAtg27 mutant was generated and gene deletion did not change growth and sporulation, whereas significantly decreased pathogenicity. FgAtg27 showed subcellular localization at pre-autophagosomal structure (PAS). After starvation induction, amount of autophagosomes in ΔFgAtg27 was significantly less than wild type and complemented strain, indicating that FgAtg27 deletion affects the autophagosome formation in F. graminearum. Meanwhile, under high Ca2+ concentration conditions, ΔFgAtg27 exhibited slowed growth, confirming that FgAtg27 also involved in F. graminearum's hyperosmotic reaction to Ca2+ concentration stress. In addition, yeast two-hybrid experiments, revealed that FgAtg27 interacts with the autophagy key protein FgAtg9. Collectively, we found that the deletion of FgAtg27 did not impact the growth phenotype of F. graminearum, whereas significantly reduced its pathogenicity and Ca2+ stress through affecting autophagic process.
Collapse
Affiliation(s)
- Yong Wen
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Mengru Wang
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Xi Liu
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Xiaohui Yin
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Shuangjun Gong
- Key Laboratory of Integrated Pest Management of Crops in Central China, Ministry of Agriculture, Hubei Key Laboratory of Crop Diseases, Insect Pests and Weeds Control, Institute of Plant Protection and Soil Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, Hubei, China
| | - Junliang Yin
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China.
| |
Collapse
|
4
|
Bui V, Liang X, Ye Y, Giang W, Tian F, Takahashi Y, Wang HG. Blocking autophagosome closure manifests the roles of mammalian Atg8-family proteins in phagophore formation and expansion during nutrient starvation. Autophagy 2024. [PMID: 39694607 DOI: 10.1080/15548627.2024.2443300] [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: 06/21/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024] Open
Abstract
Macroautophagy/autophagy, an evolutionarily conserved cellular degradation pathway, involves phagophores that sequester cytoplasmic constituents and mature into autophagosomes for subsequent lysosomal delivery. The ATG8 gene family, comprising the MAP1LC3/LC3 and GABARAP/GBR subfamilies in mammals, encodes ubiquitin-like proteins that are conjugated to phagophore membranes during autophagosome biogenesis. A central question in the field is how Atg8-family proteins are precisely involved in autophagosome formation, which remains controversial and challenging, at least in part due to the short lifespan of phagophores. In this study, we depleted the autophagosome closure regulator VPS37A to arrest autophagy at the vesicle completion step and determined the roles of mammalian Atg8-family proteins (mATG8s) in nutrient starvation-induced autophagosome biogenesis. Our investigation revealed that LC3 loss hinders phagophore formation, while GBR loss impedes both phagophore formation and expansion. The defect in membrane expansion by GBR loss appears to be attributed to compromised recruitment of ATG proteins containing an LC3-interacting region (LIR), including ULK1 and ATG3. Moreover, a combined deficiency of both LC3 and GBR subfamilies nearly completely inhibits phagophore formation, highlighting their redundant regulation of this process. Consequently, cells lacking all mATG8 members exhibit defects in downstream events such as ESCRT recruitment and autophagic flux. Collectively, these findings underscore the critical roles of mammalian Atg8-family proteins in phagophore formation and expansion during autophagy.
Collapse
Affiliation(s)
- Van Bui
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Xinwen Liang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yansheng Ye
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - William Giang
- Advanced Light Microscopy Core Facility, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Fang Tian
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yoshinori Takahashi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hong-Gang Wang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
5
|
Sankar DS, Kaeser-Pebernard S, Vionnet C, Favre S, de Oliveira Marchioro L, Pillet B, Zhou J, Stumpe M, Kovacs WJ, Kressler D, Antonioli M, Fimia GM, Dengjel J. The ULK1 effector BAG2 regulates autophagy initiation by modulating AMBRA1 localization. Cell Rep 2024; 43:114689. [PMID: 39207901 DOI: 10.1016/j.celrep.2024.114689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/15/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Autophagy initiation is regulated by the ULK1 kinase complex. To gain insights into functions of the holo-complex, we generated a deep interactome by combining affinity purification- and proximity labeling-mass spectrometry of all four complex members: ULK1, ATG13, ATG101, and RB1CC1/FIP200. Under starvation conditions, the ULK1 complex interacts with several protein and lipid kinases and phosphatases, implying the formation of a signalosome. Interestingly, several selective autophagy receptors also interact with ULK1, indicating the activation of selective autophagy pathways by nutrient starvation. One effector of the ULK1 complex is the HSC/HSP70 co-chaperone BAG2, which regulates the subcellular localization of the VPS34 lipid kinase complex member AMBRA1. Depending on the nutritional status, BAG2 has opposing roles. In growth conditions, the unphosphorylated form of BAG2 sequesters AMBRA1, attenuating autophagy induction. In starvation conditions, ULK1 phosphorylates BAG2 on Ser31, which supports the recruitment of AMBRA1 to the ER membrane, positively affecting autophagy.
Collapse
Affiliation(s)
| | | | - Christine Vionnet
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Sebastian Favre
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Lais de Oliveira Marchioro
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", 00149 Rome, Italy; Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo CEP 05508-000, Brazil
| | - Benjamin Pillet
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Jianwen Zhou
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Michael Stumpe
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Werner Josef Kovacs
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Dieter Kressler
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Manuela Antonioli
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", 00149 Rome, Italy; Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", 00149 Rome, Italy; Department of Molecular Medicine, University of Rome "Sapienza", 00185 Rome, Italy
| | - Jӧrn Dengjel
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland.
| |
Collapse
|
6
|
Leclerc NR, Dunne TM, Shrestha S, Johnson CP, Kelley JB. TOR signaling regulates GPCR levels on the plasma membrane and suppresses the Saccharomyces cerevisiae mating pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593412. [PMID: 38798445 PMCID: PMC11118302 DOI: 10.1101/2024.05.09.593412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Saccharomyces cerevisiae respond to mating pheromone through the GPCRs Ste2 and Ste3, which promote growth of a mating projection in response to ligand binding. This commitment to mating is nutritionally and energetically taxing, and so we hypothesized that the cell may suppress mating signaling during starvation. We set out to investigate negative regulators of the mating pathway in nutritionally depleted environments. Here, we report that nutrient deprivation led to loss of Ste2 from the plasma membrane. Recapitulating this effect with nitrogen starvation led us to hypothesize that it was due to TORC1 signaling. Rapamycin inhibition of TORC1 impacted membrane levels of all yeast GPCRs. Inhibition of TORC1 also dampened mating pathway output. Deletion analysis revealed that TORC1 repression leads to α-arrestin-directed CME through TORC2-Ypk1 signaling. We then set out to determine whether major downstream effectors of the TOR complexes also downregulate pathway output during mating. We found that autophagy contributes to pathway downregulation through analysis of strains lacking ATG8 . We also show that Ypk1 significantly reduced pathway output. Thus, both autophagy machinery and TORC2-Ypk1 signaling serve as attenuators of pheromone signaling during mating. Altogether, we demonstrate that the stress-responsive TOR complexes coordinate GPCR endocytosis and reduce the magnitude of pheromone signaling, in ligand-independent and ligand-dependent contexts. One Sentence Summary TOR signaling regulates the localization of all Saccharomyces cerevisiae GPCRs during starvation and suppress the mating pathway in the presence and absence of ligand.
Collapse
|
7
|
Yin Y, Ahmed N, Hassan MF, Guo K, Shakir Y, Zang J, Lyu J. Effect of Nano-selenium on Biological Mechanism of Goblet Cells of the Small Intestine Within Laying Hen. Biol Trace Elem Res 2024; 202:1699-1710. [PMID: 37454307 DOI: 10.1007/s12011-023-03770-5] [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: 04/03/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Dietary selenium intake within the normal physiological range is critical for various supporting biological functions. However, the effect of nano-selenium on biological mechanism of goblet cells associated with autophagy is largely unknown.The purpose of this study was to investigate the effect of nano-selenium on the mucosal immune-defense mechanism of goblet cells (GCs) in the small intestine of laying hens.The autophagy was determined by using specific markers. Nano-selenium-treated group of immunohistochemistry (IHC), immunofluorescence (IF), and western blotting (WB) results indicated the strong positive immune signaling of microtubule-associated light chain (LC3) within the mucosal surface of the small intestine. However, weak expression of LC3 was observed in the 3-methyladenine autophagy inhibitor (3-MA) group. IHC and IF staining results showed the opposite tendency for LC3 of sequestosome 1 (P62/SQSTM1). P62/SQSTM1 showed strong positive immune signaling within the mucosal surface of the small intestine of the 3-MAgroup, and weak immune signaling of P62/SQSTM1 in the nano-selenium-treated group. Moreover, pinpointing autophagy was involved in the mucosal production and enrichment of mucosal immunity of the GCs. The morphology and ultrastructure evidence showed that the mucus secretion of GCs was significantly increased after nano-selenium treatment confirmed by light and transmission electron microscopy. Besides that, immunostaining of IHC, IF and WB showed that autophagy enhanced the secretion of Mucin2 (Muc2) protein in nano-selenium-treated group. This work illustrates that the nano-selenium particle might enhance the mucosal immune-defense mechanism via the protective role of GCs for intestinal homeostasis through autophagy.
Collapse
Affiliation(s)
- Yongxiang Yin
- Department of Pathology, Wuxi Maternal and Child Health Care Hospital, Womens Hospital of Jiangnan University, Jiangsu, 214002, China
| | - Nisar Ahmed
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, 90150, Pakistan
| | - Mohammad Farooque Hassan
- Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand, Sindh, 67210, Pakistan
| | - Kai Guo
- Department of Pathology, Suzhou Science and Technology Town Hospital, Suzhou, 215153, China
| | - Yasmeen Shakir
- Department of Biochemistry, Hazara University, Mansehra, 21300, Pakistan
| | - Jia Zang
- Department of Laboratory Medicine, Wuxi Maternal and Child Health Care Hospital, Womens Hospital of Jiangnan University, Jiangsu, 214002, China.
| | - Jue Lyu
- Department of Laboratory Medicine, Wuxi No.2 Peoples Hospital, Jiangnan University Medical Center, Jiangsu, 214002, China.
| |
Collapse
|
8
|
Yen CC, Hsu CM, Jiang PL, Jauh GY. Dynamic organelle changes and autophagic processes in lily pollen germination. BOTANICAL STUDIES 2024; 65:5. [PMID: 38273136 PMCID: PMC10811312 DOI: 10.1186/s40529-024-00410-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
Pollen germination is a crucial process in the life cycle of flowering plants, signifying the transition of quiescent pollen grains into active growth. This study delves into the dynamic changes within organelles and the pivotal role of autophagy during lily pollen germination. Initially, mature pollen grains harbor undifferentiated organelles, including amyloplasts, mitochondria, and the Golgi apparatus. However, germination unveils remarkable transformations, such as the redifferentiation of amyloplasts accompanied by starch granule accumulation. We investigate the self-sustained nature of amylogenesis during germination, shedding light on its association with osmotic pressure. Employing BODIPY 493/503 staining, we tracked lipid body distribution throughout pollen germination, both with or without autophagy inhibitors (3-MA, NEM). Typically, lipid bodies undergo polarized movement from pollen grains into elongating pollen tubes, a process crucial for directional growth. Inhibiting autophagy disrupted this essential lipid body redistribution, underscoring the interaction between autophagy and lipid body dynamics. Notably, the presence of tubular endoplasmic reticulum (ER)-like structures associated with developing amyloplasts and lipid bodies implies their participation in autophagy. Starch granules, lipid bodies, and membrane remnants observed within vacuoles further reinforce the involvement of autophagic processes. Among the autophagy inhibitors, particularly BFA, significantly impede germination and growth, thereby affecting Golgi morphology. Immunogold labeling substantiates the pivotal role of the ER in forming autophagosome-like compartments and protein localization. Our proposed speculative model of pollen germination encompasses proplastid differentiation and autophagosome formation. This study advances our understanding of organelle dynamics and autophagy during pollen germination, providing valuable insights into the realm of plant reproductive physiology.
Collapse
Affiliation(s)
- Chih-Chung Yen
- Institute of Plant and Microbial Biology, Academia Sinica, 11529, Taipei, Taiwan, ROC
| | - Chia-Mei Hsu
- Institute of Plant and Microbial Biology, Academia Sinica, 11529, Taipei, Taiwan, ROC
| | - Pei-Luen Jiang
- Department of Biotechnology, National Formosa University, Huwei Township, Yulin County, Taiwan.
| | - Guang-Yuh Jauh
- Institute of Plant and Microbial Biology, Academia Sinica, 11529, Taipei, Taiwan, ROC.
- Molecular and Biological Agricultural Sciences, International Graduate Program, National Chung-Hsing University, Academia Sinica, Taipei, Taiwan.
- Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan.
| |
Collapse
|
9
|
Abeliovich H. Mitophagy in yeast: known unknowns and unknown unknowns. Biochem J 2023; 480:1639-1657. [PMID: 37850532 PMCID: PMC10586778 DOI: 10.1042/bcj20230279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/06/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023]
Abstract
Mitophagy, the autophagic breakdown of mitochondria, is observed in eukaryotic cells under various different physiological circumstances. These can be broadly categorized into two types: mitophagy related to quality control events and mitophagy induced during developmental transitions. Quality control mitophagy involves the lysosomal or vacuolar degradation of malfunctioning or superfluous mitochondria within lysosomes or vacuoles, and this is thought to serve as a vital maintenance function in respiring eukaryotic cells. It plays a crucial role in maintaining physiological balance, and its disruption has been associated with the progression of late-onset diseases. Developmentally induced mitophagy has been reported in the differentiation of metazoan tissues which undergo metabolic shifts upon developmental transitions, such as in the differentiation of red blood cells and muscle cells. Although the mechanistic studies of mitophagy in mammalian cells were initiated after the initial mechanistic findings in Saccharomyces cerevisiae, our current understanding of the physiological role of mitophagy in yeast remains more limited, despite the presence of better-defined assays and tools. In this review, I present my perspective on our present knowledge of mitophagy in yeast, focusing on physiological and mechanistic aspects. I aim to focus on areas where our understanding is still incomplete, such as the role of mitochondrial dynamics and the phenomenon of protein-level selectivity.
Collapse
Affiliation(s)
- Hagai Abeliovich
- Institute of Biochemistry, Food Science and Nutrition, Hebrew University of Jerusalem, 1 Hankin St, Rehovot 7610001, Israel
| |
Collapse
|
10
|
Shi X, Wu W, Feng Z, Fan P, Shi R, Zhang X. MARCH7-mediated ubiquitination decreases the solubility of ATG14 to inhibit autophagy. Cell Rep 2023; 42:113045. [PMID: 37632749 DOI: 10.1016/j.celrep.2023.113045] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/07/2023] [Accepted: 08/11/2023] [Indexed: 08/28/2023] Open
Abstract
Autophagy is a fundamental biological process critical to all eukaryotic cellular life. Although autophagy has been increasingly studied, how its process is precisely coordinated remains an open question. ATG14 (ATG14L/Barkor) is known to play a crucial role in both autophagosome formation and autophagosome-lysosome fusion. However, how ATG14 is regulated, especially at the post-translation level, is still not clear. Here, we report that MARCH7 (membrane-associated ring-CH-type finger 7), an E3 ubiquitin ligase, inhibits autophagy by ubiquitinating ATG14. MARCH7 significantly promotes K6-, K11-, and K63-linked mixed polyubiquitination on ATG14, triggering the aggregation of ATG14 and reducing its solubility in cells. Functionally, we find that MARCH7 depletion decreases the number of aggresome-like induced structures (ALISs). Mechanistically, we show that ubiquitinated ATG14 has fewer interactions with STX17, leading to the inhibition of autophagy flux. Collectively, our study reveals a mechanism in regulating autophagy and suggests a potential strategy for the treatment of autophagy-related diseases.
Collapse
Affiliation(s)
- Xue Shi
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Lineage and Development, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenfeng Wu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510530, China
| | - Zhenhuan Feng
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Lineage and Development, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiyang Fan
- SanQuan College, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Ruona Shi
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Lineage and Development, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Xiaofei Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Lineage and Development, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510530, China.
| |
Collapse
|
11
|
Su P, Chen JG, Tang DH. Exercise against nonalcoholic fatty liver disease: Possible role and mechanism of lipophagy. Life Sci 2023; 327:121837. [PMID: 37301321 DOI: 10.1016/j.lfs.2023.121837] [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: 04/13/2023] [Revised: 05/25/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common causes of chronic liver disease worldwide. NAFLD is prevalent in about 30% of people worldwide. The lack of physical activity is considered as one of the risks for NAFLD, and approximately one-third of NAFLD patients hardly engage in physical activity. It is acknowledged that exercise is one of the optimal non-pharmacological methods for preventing and treating NAFLD. Different forms of exercise such as aerobic exercise, resistance exercise and even simply physical activity in a higher level can be beneficial in reducing liver lipid accumulation and disease progression for NAFLD patients. In NAFLD patients, exercise is helpful in lowering steatosis and enhancing liver function. The mechanisms underlying the prevention and treatment of NAFLD by exercise are various and complex. Current studies on the mechanisms have focused on the pro-lipolytic, anti-inflammatory, and antioxidant and lipophagy. Promotion of lipophagy is regarded as an important mechanism for prevention and improvement of NAFLD by exercise. Recent studies have investigated the above mechanism, yet the potential mechanism has not been completely elucidated. Thus, in this review, we cover the recent advances of exercise-promoted lipophagy in NAFLD treatment and prevention. Furthermore, given the fact that exercise activates SIRT1, we discuss the possible regulatory mechanisms of lipophagy by SIRT1 during exercise. These mechanisms need to be verified by further experimental studies.
Collapse
Affiliation(s)
- Pei Su
- Department of College of P.E. and Sport, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing 100875, People's Republic of China.
| | - Jian-Gang Chen
- Department of College of P.E. and Sport, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing 100875, People's Republic of China.
| | - Dong-Hui Tang
- Department of College of P.E. and Sport, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing 100875, People's Republic of China.
| |
Collapse
|
12
|
Hua X, Liu X, Zhu Q, Liu Y, Zhou S, Huang P, Li Q, Liu S. Three-Dimensional Microfluidic Chip for Efficient Capture of Secretory Autophagosomes and Sensitive Detection of Their Surface Proteins. Anal Chem 2022; 94:8489-8496. [DOI: 10.1021/acs.analchem.2c01419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Xin Hua
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Key Laboratory of Environmental Medicine Engineering, Ministry of Education, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Xi Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Key Laboratory of Environmental Medicine Engineering, Ministry of Education, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Qian Zhu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Key Laboratory of Environmental Medicine Engineering, Ministry of Education, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yu Liu
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu 610068, Sichuan, China
| | - Sisi Zhou
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Key Laboratory of Environmental Medicine Engineering, Ministry of Education, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Puzhen Huang
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Key Laboratory of Environmental Medicine Engineering, Ministry of Education, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Quan Li
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu 610068, Sichuan, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Key Laboratory of Environmental Medicine Engineering, Ministry of Education, State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
13
|
Nguyen TN, Lazarou M. A unifying model for the role of the ATG8 system in autophagy. J Cell Sci 2022; 135:275570. [PMID: 35665638 DOI: 10.1242/jcs.258997] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/13/2022] [Indexed: 11/20/2022] Open
Abstract
The formation of autophagosomes and their fusion with lysosomes are key events that underpin autophagic degradation of cargoes. The core ATG8 system, which consists of the ATG8 family of ubiquitin-like proteins and the machineries that conjugate them onto autophagosomal membranes, are among the most-studied autophagy components. Despite the research focus on the core ATG8 system, there are conflicting reports regarding its essential roles in autophagy. Here, we reconcile prior observations of the core ATG8 system into a unifying model of their function that aims to consider apparently conflicting discoveries. Bypass pathways of autophagy that function independently of the core ATG8 system are also discussed.
Collapse
Affiliation(s)
- Thanh Ngoc Nguyen
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD 20185, USA
| | - Michael Lazarou
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD 20185, USA.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3068, Australia
| |
Collapse
|
14
|
Propofol Upregulates MicroRNA-30b to Inhibit Excessive Autophagy and Apoptosis and Attenuates Ischemia/Reperfusion Injury In Vitro and in Patients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2109891. [PMID: 35401922 PMCID: PMC8986434 DOI: 10.1155/2022/2109891] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/24/2021] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Evidence reveals that propofol protects cells via suppressing excessive autophagy induced by hypoxia/reoxygenation (H/R). Previously, we found in a genome-wide microRNA profile analysis that several autophagy-related microRNAs were significantly altered during the process of H/R in the presence or absence of propofol posthypoxia treatment (P-PostH), but how these microRNAs work in P-PostH is still largely unknown. Here, we found that one of these microRNAs, microRNA-30b (miR-30b), in human umbilical vein endothelial cells (HUVECs) was downregulated by H/R treatment but significantly upregulated by 100 M propofol after H/R treatment. miR-30b showed similar changes in open heart surgery patients. By dual-luciferase assay, we found that Beclin-1 is the direct target of miR-30b. This conclusion was also supported by knockdown or overexpression of miR-30b. Further studies showed that miR-30b inhibited H/R-induced autophagy activation. Overexpression or knockdown of miR-30b regulated autophagy-related protein gene expression in vitro. To clarify the specific role of propofol in the inhibition of autophagy and distinguish the induction of autophagy from the damage of autophagy flux, we used bafilomycin A1. LC3-II levels were decreased in the group treated with propofol combined with bafilomycin A1 compared with the group treated with bafilomycin A1 alone after hypoxia and reoxygenation. Moreover, HUVECs transfected with Ad-mCherry-GFP-LC3b confirmed the inhibitory effect of miR-30b on autophagy flux. Finally, we found that miR-30b is able to increase the cellular viability under the H/R condition, partially mimicking the protective effect of propofol which suppressed autophagy via enhancing miR-30b and targeting Beclin-1. Therefore, we concluded that propofol upregulates miR-30b to repress excessive autophagy via targeting Beclin-1 under H/R condition. Thus, our results revealed a novel mechanism of the protective role of propofol during anesthesia. Clinical Trial Registration Number. This trial is registered with ChiCTR-IPR-14005470. The name of the trial register: Propofol Upregulates MicroRNA-30b to Repress Beclin-1 and Inhibits Excessive Autophagy and Apoptosis.
Collapse
|
15
|
Meyer MD, Winzeler J, Taylor SM, Kilgore A, Edicha K, Chitwood C, Spearin Z, Silvia SKNR, Chakraborty R, Smith JE, Kennedy B, Zois C, Cawthon H, Gilruth M, Backues SK. Mapping Critical Residues in ATG11's Coiled-Coil 2 Domain that Block Multiple Interactions and Disrupt Selective Autophagy. Front Cell Dev Biol 2022; 9:775364. [PMID: 35118068 PMCID: PMC8805157 DOI: 10.3389/fcell.2021.775364] [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: 09/13/2021] [Accepted: 12/23/2021] [Indexed: 11/26/2022] Open
Abstract
Selective autophagy is a conserved subcellular process that maintains the health of eukaryotic cells by targeting damaged or toxic cytoplasmic components to the vacuole/lysosome for degradation. A key player in the initiation of selective autophagy in S. Cerevisiae (baker's yeast) is a large adapter protein called Atg11. Atg11 has multiple predicted coiled-coil domains and intrinsically disordered regions, is known to dimerize, and binds and organizes other essential components of the autophagosome formation machinery, including Atg1 and Atg9. We performed systematic directed mutagenesis on the coiled-coil 2 domain of Atg11 in order to map which residues were required for its structure and function. Using yeast-2-hybrid and coimmunoprecipitation, we found only three residues to be critical: I562, Y565, and I569. Mutation of any of these, but especially Y565, could interfere with Atg11 dimerization and block its interaction with Atg1 and Atg9, thereby inactivating selective autophagy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Steven K. Backues
- Department of Chemistry, Eastern Michigan University, Ypsilanti, MI, United States
| |
Collapse
|
16
|
Ramser A, Greene E, Alrubaye AA, Wideman R, Dridi S. Role of Autophagy Machinery Dysregulation in Bacterial Chondronecrosis with Osteomyelitis (BCO). Poult Sci 2022; 101:101750. [PMID: 35278754 PMCID: PMC8914211 DOI: 10.1016/j.psj.2022.101750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/27/2021] [Accepted: 01/23/2022] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a cell survival and homeostasis mechanism involving lysosomal degradation of cellular components and foreign bodies. It plays a role in bone homeostasis, skeletal diseases, and bacterial infections as both a cell-survival or cell-death pathway. This study sought to determine if autophagy played a role in bacterial chondronecrosis with osteomyelitis (BCO). BCO is a prominent cause of lameness in modern broilers and results from bacterial infection of mechanically stressed leg bone growth plates. The protein and gene expression of key autophagy machinery was analyzed in both normal and BCO-affected broilers using real-time qPCR and immunoblot, respectively. Gene expression showed a significant downregulation of key target signatures involved in every stage of autophagy in BCO-affected bone, such as ATG13, SQSTM1 (p62), ATG9B, ATG16L, ATG12, LC3C, and RAB7A. Additionally, protein expression for LC3 was also significantly lower in BCO. An in vitro study using human fetal osteoblast cells challenged with BCO isolate, Staphylococcus agnetis 908, showed a similar dysregulation of autophagy machinery along with a significant decrease in cell viability. When autophagy was inhibited via 3-methyladenine or chloroquine, comparable decreases in cell viability were seen along with dysregulation of autophagy machinery. Together, these results are the first to implicate autophagy machinery dysregulation in the pathology of BCO.
Collapse
|
17
|
Padman BS, Lazarou M. Immunofluorescence-Based Measurement of Autophagosome Formation During Mitophagy. Methods Mol Biol 2022; 2445:207-226. [PMID: 34972994 DOI: 10.1007/978-1-0716-2071-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Damaged, dysfunctional, or excess mitochondria are removed from cells via a selective form of macroautophagy termed mitophagy. The clearance of mitochondria during mitophagy is mediated by double-membrane vesicles called autophagosomes, which encapsulate mitochondria that have been tagged for mitophagic removal before delivering them to lysosomes for degradation. A variety of different mitophagy pathways exist that differ in their mechanisms of initiation but share a common pathway of autophagosome formation. Autophagosome biogenesis is regulated by a number of autophagy factors which translocate from the cytosol to spatially defined focal points (foci) on the mitochondrial surface after mitophagy has been initiated. The functional analysis of autophagosome biogenesis requires the use of microscopy-based techniques which assess the recruitment of autophagy factors to mitophagic foci representing autophagosome formation sites. Here, we describe a routine method for the quantitative 3D analysis of mitophagic foci in PINK1/Parkin mitophagy immunofluorescence samples through the application of object-based image analysis (OBIA) to 3D confocal imaging datasets. The approach enables unbiased high-throughput characterisation of autophagosome biogenesis during mitophagy.
Collapse
Affiliation(s)
- Benjamin S Padman
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth, WA, Australia.
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
18
|
Lei Y, Zhang X, Xu Q, Liu S, Li C, Jiang H, Lin H, Kong E, Liu J, Qi S, Li H, Xu W, Lu K. Autophagic elimination of ribosomes during spermiogenesis provides energy for flagellar motility. Dev Cell 2021; 56:2313-2328.e7. [PMID: 34428398 DOI: 10.1016/j.devcel.2021.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 05/17/2021] [Accepted: 07/23/2021] [Indexed: 02/05/2023]
Abstract
How autophagy initiation is regulated and what the functional significance of this regulation is are unknown. Here, we characterized the role of yeast Vac8 in autophagy initiation through recruitment of PIK3C3-C1 to the phagophore assembly site (PAS). This recruitment is dependent on the palmitoylation of Vac8 and on its middle ARM domains for binding PIK3C3-C1. Vac8-mediated anchoring of PIK3C3-C1 promotes PtdIns3P generation at the PAS and recruitment of the PtdIns3P binding protein Atg18-Atg2. The mouse homolog of Vac8, ARMC3, is conserved and functions in autophagy in mouse testes. Mice lacking ARMC3 have normal viability but show complete male infertility. Proteomic analysis indicated that the autophagic degradation of cytosolic ribosomes was blocked in ARMC3-deficient spermatids, which caused low energy levels of mitochondria and motionless flagella. These studies uncovered a function of Vac8/ARMC3 in PtdIns3-kinase anchoring at the PAS and its physical significance in mammalian spermatogenesis with a germ tissue-specific autophagic function.
Collapse
Affiliation(s)
- Yuqing Lei
- Department of Pathology, West China Second University Hospital, State Key Laboratory of Biotherapy, and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Xueguang Zhang
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Qingjia Xu
- Department of Neurology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiyan Liu
- Department of Neurology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunxia Li
- Department of Neurology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China; Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Beijing 100191, China; Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China
| | - Eryan Kong
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang 453003, China
| | - Jiaming Liu
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiqian Qi
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huihui Li
- Department of Pathology, West China Second University Hospital, State Key Laboratory of Biotherapy, and Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu 610041, China.
| | - Wenming Xu
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Kefeng Lu
- Department of Neurology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
19
|
Barz S, Kriegenburg F, Sánchez-Martín P, Kraft C. Small but mighty: Atg8s and Rabs in membrane dynamics during autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119064. [PMID: 34048862 PMCID: PMC8261831 DOI: 10.1016/j.bbamcr.2021.119064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/04/2021] [Accepted: 05/21/2021] [Indexed: 11/17/2022]
Abstract
Autophagy is a degradative pathway during which autophagosomes are formed that enwrap cytosolic material destined for turnover within the lytic compartment. Autophagosome biogenesis requires controlled lipid and membrane rearrangements to allow the formation of an autophagosomal seed and its subsequent elongation into a fully closed and fusion-competent double membrane vesicle. Different membrane remodeling events are required, which are orchestrated by the distinct autophagy machinery. An important player among these autophagy proteins is the small lipid-modifier Atg8. Atg8 proteins facilitate various aspects of autophagosome formation and serve as a binding platform for autophagy factors. Also Rab GTPases have been implicated in autophagosome biogenesis. As Atg8 proteins interact with several Rab GTPase regulators, they provide a possible link between autophagy progression and Rab GTPase activity. Here, we review central aspects in membrane dynamics during autophagosome biogenesis with a focus on Atg8 proteins and selected Rab GTPases.
Collapse
Affiliation(s)
- Saskia Barz
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Franziska Kriegenburg
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Pablo Sánchez-Martín
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Claudine Kraft
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
20
|
Nguyen TN, Padman BS, Zellner S, Khuu G, Uoselis L, Lam WK, Skulsuppaisarn M, Lindblom RSJ, Watts EM, Behrends C, Lazarou M. ATG4 family proteins drive phagophore growth independently of the LC3/GABARAP lipidation system. Mol Cell 2021; 81:2013-2030.e9. [PMID: 33773106 DOI: 10.1016/j.molcel.2021.03.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 10/20/2020] [Accepted: 02/25/2021] [Indexed: 02/08/2023]
Abstract
The sequestration of damaged mitochondria within double-membrane structures termed autophagosomes is a key step of PINK1/Parkin mitophagy. The ATG4 family of proteases are thought to regulate autophagosome formation exclusively by processing the ubiquitin-like ATG8 family (LC3/GABARAPs). We discover that human ATG4s promote autophagosome formation independently of their protease activity and of ATG8 family processing. ATG4 proximity networks reveal a role for ATG4s and their proximity partners, including the immune-disease protein LRBA, in ATG9A vesicle trafficking to mitochondria. Artificial intelligence-directed 3D electron microscopy of phagophores shows that ATG4s promote phagophore-ER contacts during the lipid-transfer phase of autophagosome formation. We also show that ATG8 removal during autophagosome maturation does not depend on ATG4 activity. Instead, ATG4s can disassemble ATG8-protein conjugates, revealing a role for ATG4s as deubiquitinating-like enzymes. These findings establish non-canonical roles of the ATG4 family beyond the ATG8 lipidation axis and provide an AI-driven framework for rapid 3D electron microscopy.
Collapse
Affiliation(s)
- Thanh Ngoc Nguyen
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
| | - Benjamin Scott Padman
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Susanne Zellner
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Grace Khuu
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Louise Uoselis
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Wai Kit Lam
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Marvin Skulsuppaisarn
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Runa S J Lindblom
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Emily M Watts
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Christian Behrends
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
| |
Collapse
|
21
|
Melia TJ, Lystad AH, Simonsen A. Autophagosome biogenesis: From membrane growth to closure. J Cell Biol 2021; 219:151729. [PMID: 32357219 PMCID: PMC7265318 DOI: 10.1083/jcb.202002085] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
Autophagosome biogenesis involves de novo formation of a membrane that elongates to sequester cytoplasmic cargo and closes to form a double-membrane vesicle (an autophagosome). This process has remained enigmatic since its initial discovery >50 yr ago, but our understanding of the mechanisms involved in autophagosome biogenesis has increased substantially during the last 20 yr. Several key questions do remain open, however, including, What determines the site of autophagosome nucleation? What is the origin and lipid composition of the autophagosome membrane? How is cargo sequestration regulated under nonselective and selective types of autophagy? This review provides key insight into the core molecular mechanisms underlying autophagosome biogenesis, with a specific emphasis on membrane modeling events, and highlights recent conceptual advances in the field.
Collapse
Affiliation(s)
- Thomas J Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Alf H Lystad
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Fu C, Tian G, Duan J, Liu K, Zhang C, Yan W, Wang Y. Therapeutic Antitumor Efficacy of Cancer Stem Cell-Derived DRibble Vaccine on Colorectal Carcinoma. Int J Med Sci 2021; 18:3249-3260. [PMID: 34400894 PMCID: PMC8364449 DOI: 10.7150/ijms.61510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC)-based immunotherapy has been a promising strategy for colon cancer therapy, but the efficacy of dendritic cell vaccines is in part limited by immunogenicity of loaded antigens. In this study, we aimed to identify a putative tumor antigen that can generate or enhance anti-tumor immune responses against colon cancer. CD44+ colon cancer stem cells (CCSCs) were isolated from mouse colorectal carcinoma CT-26 cell cultures and induced to form defective ribosomal products-containing autophagosome-rich blebs (DRibbles) by treatment with rapamycin, bortezomib, and ammonium chloride. DRibbles were characterized by western blot and transmission electron microscopy. DCs generated from the mice bone marrow monocytes were cocultured with DRibbles, then surface markers of DCs were analyzed by flow cytometry. Meanwhile, the efficacy of DRibble-DCs was examined in vivo. Our results showed that CCSC-derived DRibbles upregulated CD80, CD86, major histocompatibility complex (MHC)-I, and MHC-II on DCs and induced proliferation of mouse splenic lymphocytes and CD8+ T cells. In a model of colorectal carcinoma using BALB/c mice with robust tumor growth and mortality, DC vaccine pulsed with CCSC-derived DRibbles suppressed tumor growth and extended survival. A lactate dehydrogenase test indicated a strong cytolytic activity of cytotoxic T-cells derived from mice vaccinated with CCSC-derived DRibbles against CT-26 cells. Furthermore, flow cytometry analyses showed that the percentages of IFN-γ-producing CD8+ T-cells were increased in SD-DC group compare with the other groups. These findings provide a rationale for novel immunotherapeutic anti-tumor approaches based on DRibbles derived from colon cancer stem cells.
Collapse
Affiliation(s)
- Changhao Fu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China.,Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Geer Tian
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Jinyue Duan
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Kun Liu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Chen Zhang
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Weiqun Yan
- Medical Institute of Regeneration Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
23
|
Rangarajan N, Kapoor I, Li S, Drossopoulos P, White KK, Madden VJ, Dohlman HG. Potassium starvation induces autophagy in yeast. J Biol Chem 2020; 295:14189-14202. [PMID: 32788210 DOI: 10.1074/jbc.ra120.014687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/02/2020] [Indexed: 01/04/2023] Open
Abstract
Autophagy is a conserved process that recycles cellular contents to promote survival. Although nitrogen limitation is the canonical inducer of autophagy, recent studies have revealed several other nutrients important to this process. In this study, we used a quantitative, high-throughput assay to identify potassium starvation as a new and potent inducer of autophagy in the yeast Saccharomyces cerevisiae We found that potassium-dependent autophagy requires the core pathway kinases Atg1, Atg5, and Vps34, and other components of the phosphatidylinositol 3-kinase complex. Transmission EM revealed abundant autophagosome formation in response to both stimuli. RNA-Seq indicated distinct transcriptional responses: nitrogen affects transport of ions such as copper, whereas potassium targets the organization of other cellular components. Thus, nitrogen and potassium share the ability to influence molecular supply and demand but do so in different ways. Both inputs promote catabolism through bulk autophagy, but result in distinct mechanisms of cellular remodeling and synthesis.
Collapse
Affiliation(s)
- Nambirajan Rangarajan
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ishani Kapoor
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shuang Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter Drossopoulos
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kristen K White
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Victoria J Madden
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Henrik G Dohlman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
24
|
Munzel L, Neumann P, Otto FB, Krick R, Metje-Sprink J, Kroppen B, Karedla N, Enderlein J, Meinecke M, Ficner R, Thumm M. Atg21 organizes Atg8 lipidation at the contact of the vacuole with the phagophore. Autophagy 2020; 17:1458-1478. [PMID: 32515645 DOI: 10.1080/15548627.2020.1766332] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Coupling of Atg8 to phosphatidylethanolamine is crucial for the expansion of the crescent-shaped phagophore during cargo engulfment. Atg21, a PtdIns3P-binding beta-propeller protein, scaffolds Atg8 and its E3-like complex Atg12-Atg5-Atg16 during lipidation. The crystal structure of Atg21, in complex with the Atg16 coiled-coil domain, showed its binding at the bottom side of the Atg21 beta-propeller. Our structure allowed detailed analyses of the complex formation of Atg21 with Atg16 and uncovered the orientation of the Atg16 coiled-coil domain with respect to the membrane. We further found that Atg21 was restricted to the phagophore edge, near the vacuole, known as the vacuole isolation membrane contact site (VICS). We identified a specialized vacuolar subdomain at the VICS, typical of organellar contact sites, where the membrane protein Vph1 was excluded, while Vac8 was concentrated. Furthermore, Vac8 was required for VICS formation. Our results support a specialized organellar contact involved in controlling phagophore elongation. Abbreviations: FCCS: fluorescence cross correlation spectroscopy; NVJ: nucleus-vacuole junction; PAS: phagophore assembly site; PE: phosphatidylethanolamine; PROPPIN: beta-propeller that binds phosphoinositides; PtdIns3P: phosphatidylinositol- 3-phosphate; VICS: vacuole isolation membrane contact site.
Collapse
Affiliation(s)
- Lena Munzel
- Department of Cellular Biochemistry, University Medicine, Goettingen, Germany
| | - Piotr Neumann
- Department of Molecular Structural Biology, Institute for Microbiology and Genetics, University of Goettingen, Goettingen, Germany
| | - Florian B Otto
- Department of Cellular Biochemistry, University Medicine, Goettingen, Germany
| | - Roswitha Krick
- Department of Cellular Biochemistry, University Medicine, Goettingen, Germany
| | - Janina Metje-Sprink
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Benjamin Kroppen
- Department of Cellular Biochemistry, University Medicine, Goettingen, Germany
| | - Narain Karedla
- Physics Department III, University of Goettingen, Goettingen, Germany
| | - Jörg Enderlein
- Physics Department III, University of Goettingen, Goettingen, Germany
| | - Michael Meinecke
- Department of Cellular Biochemistry, University Medicine, Goettingen, Germany
| | - Ralf Ficner
- Department of Molecular Structural Biology, Institute for Microbiology and Genetics, University of Goettingen, Goettingen, Germany
| | - Michael Thumm
- Department of Cellular Biochemistry, University Medicine, Goettingen, Germany
| |
Collapse
|
25
|
Taniguchi S, Toyoshima M, Takamatsu T, Mima J. Curvature-sensitive trans-assembly of human Atg8-family proteins in autophagy-related membrane tethering. Protein Sci 2020; 29:1387-1400. [PMID: 31960529 DOI: 10.1002/pro.3828] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/18/2022]
Abstract
In macroautophagy, de novo formation of the double membrane-bound organelles, termed autophagosomes, is essential for engulfing and sequestering the cytoplasmic contents to be degraded in the lytic compartments such as vacuoles and lysosomes. Atg8-family proteins have been known to be responsible for autophagosome formation via membrane tethering and fusion events of precursor membrane structures. Nevertheless, how Atg8 proteins act directly upon autophagosome formation still remains enigmatic. Here, to further gain molecular insights into Atg8-mediated autophagic membrane dynamics, we study the two representative human Atg8 orthologs, LC3B and GATE-16, by quantitatively evaluating their intrinsic potency to physically tether lipid membranes in a chemically defined reconstitution system using purified Atg8 proteins and synthetic liposomes. Both LC3B and GATE-16 retained the capacities to trigger efficient membrane tethering at the protein-to-lipid molar ratios ranging from 1:100 to 1:5,000. These human Atg8-mediated membrane-tethering reactions require trans-assembly between the membrane-anchored forms of LC3B and GATE-16 and can be reversibly and strictly controlled by the membrane attachment and detachment cycles. Strikingly, we further uncovered distinct membrane curvature dependences of LC3B- and GATE-16-mediated membrane tethering reactions: LC3B can drive tethering more efficiently than GATE-16 for highly curved small vesicles (e.g., 50 nm in diameter), although GATE-16 turns out to be a more potent tether than LC3B for flatter large vesicles (e.g., 200 and 400 nm in diameter). Our findings establish curvature-sensitive trans-assembly of human Atg8-family proteins in reconstituted membrane tethering, which recapitulates an essential subreaction of the biogenesis of autophagosomes in vivo.
Collapse
Affiliation(s)
- Saki Taniguchi
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | | | - Tomoyo Takamatsu
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Joji Mima
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
26
|
Rangarajan N, Gordy CL, Askew L, Bevill SM, Elston TC, Errede B, Hurst JH, Kelley JB, Sheetz JB, Suzuki SK, Valentin NH, Young E, Dohlman HG. Systematic analysis of F-box proteins reveals a new branch of the yeast mating pathway. J Biol Chem 2019; 294:14717-14731. [PMID: 31399514 DOI: 10.1074/jbc.ra119.010063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/06/2019] [Indexed: 11/06/2022] Open
Abstract
The mating pathway in yeast Saccharomyces cerevisiae has long been used to reveal new mechanisms of signal transduction. The pathway comprises a pheromone receptor, a heterotrimeric G protein, and intracellular effectors of morphogenesis and transcription. Polarized cell growth, in the direction of a potential mating partner, is accomplished by the G-protein βγ subunits and the small G-protein Cdc42. Transcription induction, needed for cell-cell fusion, is mediated by Gβγ and the mitogen-activated protein kinase (MAPK) scaffold protein Ste5. A potential third pathway is initiated by the G-protein α subunit Gpa1. Gpa1 signaling was shown previously to involve the F-box adaptor protein Dia2 and an endosomal effector protein, the phosphatidylinositol 3-kinase Vps34. Vps34 is also required for proper vacuolar sorting and autophagy. Here, using a panel of reporter assays, we demonstrate that mating pheromone stimulates vacuolar targeting of a cytoplasmic reporter protein and that this process depends on Vps34. Through a systematic analysis of F-box deletion mutants, we show that Dia2 is required to sustain pheromone-induced vacuolar targeting. We also found that other F-box proteins selectively regulate morphogenesis (Ydr306, renamed Pfu1) and transcription (Ucc1). These findings point to the existence of a new and distinct branch of the pheromone-signaling pathway, one that likely leads to vacuolar engulfment of cytoplasmic proteins and recycling of cellular contents in preparation for mating.
Collapse
Affiliation(s)
- Nambirajan Rangarajan
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Claire L Gordy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Lauren Askew
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Samantha M Bevill
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Timothy C Elston
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Beverly Errede
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Jillian H Hurst
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Joshua B Kelley
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Joshua B Sheetz
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Sara Kimiko Suzuki
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Natalie H Valentin
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Everett Young
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Henrik G Dohlman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
27
|
Han Z, Wang W, Lv X, Zong Y, Liu S, Liu Z, Wang L, Song L. ATG10 (autophagy-related 10) regulates the formation of autophagosome in the anti-virus immune response of pacific oyster (Crassostrea gigas). FISH & SHELLFISH IMMUNOLOGY 2019; 91:325-332. [PMID: 31128297 DOI: 10.1016/j.fsi.2019.05.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 06/09/2023]
Abstract
Autophagy, a highly conserved intracellular degradation system, is involved in numerous processes in vertebrate and invertebrate, such as cell survival, ageing, and immune responses. However, the detailed molecular mechanism of autophagy and its immune regulatory role in bivalves are still not well understood. In the present study, an autophagy-related protein ATG10 (designated as CgATG10) was identified from Pacific oyster Crassostrea gigas. The open reading frame of CgATG10 cDNA was of 621 bp, encoding a polypeptide of 206 amino acid residues with an Autophagy_act_C domain (from 96 to 123 amino acid), which shared high homology with that from C. virginica and Octopus bimaculoides. The mRNA transcripts of CgATG10 were widely expressed in all the tested tissues including mantle, gonad, gills, hemocytes and hepatopancreas, with the highest expression level in mantle. After the stimulation with poly (I:C), the mRNA expression level of CgATG10 in the mantle of oysters was significantly up-regulated (4.92-fold of that in Blank group, p < 0.05), and the LC3-conversion from LC3-I to LC3-II (LC3-II/LC3-I) also increased. After an additional injection of dsRNA to knock-down the expression of CgATG10 (0.33-fold and 0.10-fold compared respectively with Blank group and dsGFP group, p < 0.05), the downstream conversion of CgLC3 was inhibited significantly compared with that of the control dsGFP group, while the expression level of autophagy-initiator CgBeclin1 did not change significantly. In addition, the mRNA transcripts of interferon regulatory factor CgIRF-1 increased significantly in CgATG10-knockdown oysters at 12 h post poly (I:C) stimulation. All the results indicated that CgATG10 might participate in the immune response against poly (I:C) by regulating autophagosome formation and interferon system in oysters.
Collapse
Affiliation(s)
- Zirong Han
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
| | - Xiaojing Lv
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Yanan Zong
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Shujing Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Zhaoqun Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
28
|
Cao Y, Li Q, Liu L, Wu H, Huang F, Wang C, Lan Y, Zheng F, Xing F, Zhou Q, Li Q, Shi H, Zhang B, Wang Z, Wu X. Modafinil protects hippocampal neurons by suppressing excessive autophagy and apoptosis in mice with sleep deprivation. Br J Pharmacol 2019; 176:1282-1297. [PMID: 30767208 DOI: 10.1111/bph.14626] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/05/2018] [Accepted: 01/27/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Sleep deprivation compromises learning and memory in both humans and animals, and can be reversed by administration of modafinil, a drug promoting wakefulness. Dysfunctional autophagy increases activation of apoptotic cascades, ultimately leading to increased neuronal death, which can be alleviated by autophagy inhibitors. This study aimed to investigate the alleviative effect and mechanism of modafinil on the excessive autophagy occurring in the hippocampus of mice with deficiency of learning and memory induced by sleep deprivation. EXPERIMENTAL APPROACH The Morris water maze was used to assess the effects of modafinil on male C57BL/6Slac mice after 48-hr sleep deprivation. The HT-22 hippocampal neuronal cell line was also used. Nissl staining, transmission electron microscope, immunofluorescence, Western blot, transient transfection, and autophagy inducer were used to study the effect and mechanism of modafinil on hippocampal neurons with excessive autophagy and apoptosis. KEY RESULTS Modafinil improved learning and memory in sleep-deprived mice, associated with the inhibition of excessive autophage and apoptosis and an enhanced activation of the PI3K/Akt/mTOR/P70S6K signalling pathway in hippocampal neurons. These effects of modafinil were abolished by rapamycin. In addition, modafinil suppressed the aberrant autophagy and apoptosis induced by rapamycin and reactivated PI3K/Akt/mTOR/P70S6K signals in HT-22 cells. CONCLUSIONS AND IMPLICATIONS These results suggested that modafinil alleviated impaired learning and memory of sleep-deprived mice potentially by suppressing excessive autophagy and apoptosis of hippocampal neurons. This novel mechanism may add to our knowledge of modafinil in the clinical treatment of impaired memory caused by sleep loss.
Collapse
Affiliation(s)
- Yin Cao
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qinglin Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Lulu Liu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Changhong Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunyi Lan
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fang Zheng
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Faping Xing
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiang Zhou
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Beibei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengtao Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
29
|
High-mobility group protein N2 induces autophagy by activating AMPK/ULK1 pathway and thereby boosts UPEC proliferation within bladder epithelial cells. Biochem Biophys Res Commun 2019; 513:193-200. [PMID: 30952427 DOI: 10.1016/j.bbrc.2019.03.062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/11/2019] [Indexed: 02/05/2023]
Abstract
Urinary tract infection is one of the most common bacterial infections which is mainly caused by Escherichia coli (UPEC). Autophagy plays a key role in immune response to eliminate invading pathogens. Exploring the effect of autophagy on UPEC infection and the molecular mechanisms will be benefit for the treatment of urinary tract infection. High-mobility group protein N2 (HMGN2), a highly conserved nuclear protein and an antibacterial peptide, has been associated with bacterial infection induced immune response; however, whether this function is due to the regulation of autophagy remains unclear. In this study, we demonstrate for the first time that HMGN2 is upregulated in UPEC infection of bladder epithelial cell line 5637 (BEC 5637). Furthermore, HMGN2 enhances autophagy in BEC 5637 via activation of AMPK and ULK1, whereas UPEC suppresses autophagy. In addition, the enhanced autophagy activity by HMGN2 overexpression or rapamycin boosts the proliferation of UPEC J96 in BEC 5637. In summary, our data indicate that HMGN2 activates autophagy via AMPK/ULK1 pathway which can be utilized by UPEC J96 for their proliferation within bladder epithelial cells.
Collapse
|
30
|
Loeffler DA. Influence of Normal Aging on Brain Autophagy: A Complex Scenario. Front Aging Neurosci 2019; 11:49. [PMID: 30914945 PMCID: PMC6421305 DOI: 10.3389/fnagi.2019.00049] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Misfolded proteins are pathological findings in some chronic neurodegenerative disorders including Alzheimer's, Parkinson's, and Huntington's diseases. Aging is a major risk factor for these disorders, suggesting that the mechanisms responsible for clearing misfolded proteins from the brain, the ubiquitin-proteasome system and the autophagy-lysosomal pathway, may decline with age. Although autophagic mechanisms have been found to decrease with age in many experimental models, whether they do so in the brain is unclear. This review examines the literature with regard to age-associated changes in macroautophagy and chaperone-mediated autophagy (CMA) in the central nervous system (CNS). Beclin 1, LC3-II, and the LC3-II/LC3-I ratio have frequently been used to examine changes in macroautophagic activity, while lamp2a and HSPA8 (also known as hsc70) have been used to measure CMA activity. Three gene expression analyses found evidence for an age-related downregulation of macroautophagy in human brain, but no published studies were found of age-related changes in CMA in human brain, although cerebrospinal fluid concentrations of HSPA8 were reported to decrease with age. Most studies of age-related changes in brain autophagy in experimental animals have found age-related declines in macroautophagy, and macroautophagy is necessary for normal lifespan in Caenorhabditis elegans, Drosophila, and mice. However, the few studies of age-related changes in brain CMA in experimental animals have produced conflicting results. Investigations of the influence of aging on macroautophagy in experimental animals in systems other than the CNS have generally found an age-related decrease in Beclin 1, but conflicting results for LC3-II and the LC3-II/LC3-I ratio, while CMA decreases with age in most models. CONCLUSION: while indirect evidence suggests that brain autophagy may decrease with normal aging, this issue has not been investigated sufficiently, particularly in human brain. Measuring autophagic activity in the brain can be challenging because of differences in basal autophagic activity between experimental models, and the inability to include lysosomal inhibitors when measuring the LC3-II/LC3-I ratio in postmortem specimens. If autophagy does decrease in the brain with aging, then pharmacological interventions and/or lifestyle alterations to slow this decline could reduce the risk of developing age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- David A Loeffler
- Beaumont Research Institute, Department of Neurology, Beaumont Health, Royal Oak, MI, United States
| |
Collapse
|
31
|
LC3/GABARAPs drive ubiquitin-independent recruitment of Optineurin and NDP52 to amplify mitophagy. Nat Commun 2019; 10:408. [PMID: 30679426 PMCID: PMC6345886 DOI: 10.1038/s41467-019-08335-6] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 12/21/2018] [Indexed: 12/17/2022] Open
Abstract
Current models of selective autophagy dictate that autophagy receptors, including Optineurin and NDP52, link cargo to autophagosomal membranes. This is thought to occur via autophagy receptor binding to Atg8 homologs (LC3/GABARAPs) through an LC3 interacting region (LIR). The LIR motif within autophagy receptors is therefore widely recognised as being essential for selective sequestration of cargo. Here we show that the LIR motif within OPTN and NDP52 is dispensable for Atg8 recruitment and selectivity during PINK1/Parkin mitophagy. Instead, Atg8s play a critical role in mediating ubiquitin-independent recruitment of OPTN and NDP52 to growing phagophore membranes via the LIR motif. The additional recruitment of OPTN and NDP52 amplifies mitophagy through an Atg8-dependent positive feedback loop. Rather than functioning in selectivity, our discovery of a role for the LIR motif in mitophagy amplification points toward a general mechanism by which Atg8s can recruit autophagy factors to drive autophagosome growth and amplify selective autophagy.
Collapse
|
32
|
PP2C phosphatases promote autophagy by dephosphorylation of the Atg1 complex. Proc Natl Acad Sci U S A 2019; 116:1613-1620. [PMID: 30655342 DOI: 10.1073/pnas.1817078116] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Macroautophagy is orchestrated by the Atg1-Atg13 complex in budding yeast. Under nutrient-rich conditions, Atg13 is maintained in a hyperphosphorylated state by the TORC1 kinase. After nutrient starvation, Atg13 is dephosphorylated, triggering Atg1 kinase activity and macroautophagy induction. The phosphatases that dephosphorylate Atg13 remain uncharacterized. Here, we show that two redundant PP2C phosphatases, Ptc2 and Ptc3, regulate macroautophagy by dephosphorylating Atg13 and Atg1. In the absence of these phosphatases, starvation-induced macroautophagy and the cytoplasm-to-vacuole targeting pathway are inhibited, and the recruitment of the essential autophagy machinery to the phagophore assembly site is impaired. Expressing a genomic ATG13 -8SA allele lacking key TORC1 phosphorylation sites partially bypasses the macroautophagy defect in ptc2Δ ptc3Δ strains. Moreover, Ptc2 and Ptc3 interact with the Atg1-Atg13 complex. Taken together, these results suggest that PP2C-type phosphatases promote macroautophagy by regulating the Atg1 complex.
Collapse
|
33
|
Norizuki T, Kanazawa T, Minamino N, Tsukaya H, Ueda T. Marchantia polymorpha, a New Model Plant for Autophagy Studies. FRONTIERS IN PLANT SCIENCE 2019; 10:935. [PMID: 31379911 PMCID: PMC6652269 DOI: 10.3389/fpls.2019.00935] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/03/2019] [Indexed: 05/18/2023]
Abstract
Autophagy is a catabolic process for bulk and selective degradation of cytoplasmic components in the vacuole/lysosome. In Saccharomyces cerevisiae, ATG genes were identified as essential genes for autophagy, and most ATG genes are highly conserved among eukaryotes, including plants. Although reverse genetic analyses have revealed that autophagy is involved in responses to abiotic and biotic stresses in land plants, our knowledge of its molecular mechanism remains limited. This limitation is partly because of the multiplication of some ATG genes, including ATG8, in widely used model plants such as Arabidopsis thaliana, which adds complexity to functional studies. Furthermore, due to limited information on the composition and functions of the ATG genes in basal land plants and charophytes, it remains unclear whether multiplication of ATG genes is associated with neofunctionalization of these genes. To gain insight into the diversification of ATG genes during plant evolution, we compared the composition of ATG genes in plants with a special focus on a liverwort and two charophytes, which have not previously been analyzed. Our results showed that the liverwort Marchantia polymorpha and the charophytes Klebsormidium nitens and Chara braunii harbor fundamental sets of ATG genes with low redundancy compared with those of A. thaliana and the moss Physcomitrella patens, suggesting that multiplication of ATG genes occurred during land plant evolution. We also attempted to establish an experimental system for analyzing autophagy in M. polymorpha. We generated transgenic plants expressing fluorescently tagged MpATG8 to observe its dynamics in M. polymorpha and produced autophagy-defective mutants by genome editing using the CRISPR/Cas9 system. These tools allowed us to demonstrate that MpATG8 is transported into the vacuole in an MpATG2-, MpATG5-, and MpATG7-dependent manner, suggesting that fluorescently tagged MpATG8 can be used as an autophagosome marker in M. polymorpha. M. polymorpha can provide a powerful system for studying the mechanisms and evolution of autophagy in plants.
Collapse
Affiliation(s)
- Takuya Norizuki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Division of Cellular Dynamics, National Institute for Basic Biology, Okazaki, Japan
| | - Takehiko Kanazawa
- Division of Cellular Dynamics, National Institute for Basic Biology, Okazaki, Japan
- Department of Basic Biology, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Naoki Minamino
- Division of Cellular Dynamics, National Institute for Basic Biology, Okazaki, Japan
| | - Hirokazu Tsukaya
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Takashi Ueda
- Division of Cellular Dynamics, National Institute for Basic Biology, Okazaki, Japan
- Department of Basic Biology, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
- *Correspondence: Takashi Ueda,
| |
Collapse
|
34
|
Moparthi SB, Wollert T. Reconstruction of destruction – in vitro reconstitution methods in autophagy research. J Cell Sci 2018; 132:132/4/jcs223792. [DOI: 10.1242/jcs.223792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
ABSTRACT
Autophagy is one of the most elaborative membrane remodeling systems in eukaryotic cells. Its major function is to recycle cytoplasmic material by delivering it to lysosomes for degradation. To achieve this, a membrane cisterna is formed that gradually captures cargo such as organelles or protein aggregates. The diversity of cargo requires autophagy to be highly versatile to adapt the shape of the phagophore to its substrate. Upon closure of the phagophore, a double-membrane-surrounded autophagosome is formed that eventually fuses with lysosomes. In response to environmental cues such as cytotoxicity or starvation, bulk cytoplasm can be captured and delivered to lysosomes. Autophagy thus supports cellular survival under adverse conditions. During the past decades, groundbreaking genetic and cell biological studies have identified the core machinery involved in the process. In this Review, we are focusing on in vitro reconstitution approaches to decipher the details and spatiotemporal control of autophagy, and how such studies contributed to our current understanding of the pathways in yeast and mammals. We highlight studies that revealed the function of the autophagy machinery at a molecular level with respect to its capacity to remodel membranes.
Collapse
Affiliation(s)
- Satish Babu Moparthi
- Membrane Biochemistry and Transport, Institute Pasteur, 28 rue du Dr Roux, 75015 Paris, France
| | - Thomas Wollert
- Membrane Biochemistry and Transport, Institute Pasteur, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
35
|
Ye X, Zhou XJ, Zhang H. Exploring the Role of Autophagy-Related Gene 5 ( ATG5) Yields Important Insights Into Autophagy in Autoimmune/Autoinflammatory Diseases. Front Immunol 2018; 9:2334. [PMID: 30386331 PMCID: PMC6199349 DOI: 10.3389/fimmu.2018.02334] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 09/19/2018] [Indexed: 12/31/2022] Open
Abstract
Autophagy is a highly conserved process that degrades certain intracellular contents in both physiological and pathological conditions. Autophagy-related proteins (ATG) are key players in this pathway, among which ATG5 is indispensable in both canonical and non-canonical autophagy. Recent studies demonstrate that ATG5 modulates the immune system and crosstalks with apoptosis. However, our knowledge of the pathogenesis and regulatory mechanisms of autophagy in various immune related diseases is lacking. Thus, a deeper understanding of ATG5's role in the autophagy mechanism may shed light on the link between autophagy and the immune response, and lead to the development of new therapies for autoimmune diseases and autoinflammatory diseases. In this focused review, we discuss the latest insights into the role of ATG5 in autoimmunity. Although these studies are at a relatively early stage, ATG5 may eventually come to be regarded as a “guardian of immune integrity.” Notably, accumulating evidence indicates that other ATG genes may have similar functions.
Collapse
Affiliation(s)
- Xin Ye
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
36
|
Kauffman KJ, Yu S, Jin J, Mugo B, Nguyen N, O'Brien A, Nag S, Lystad AH, Melia TJ. Delipidation of mammalian Atg8-family proteins by each of the four ATG4 proteases. Autophagy 2018; 14:992-1010. [PMID: 29458288 DOI: 10.1080/15548627.2018.1437341] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
During macroautophagy/autophagy, mammalian Atg8-family proteins undergo 2 proteolytic processing events. The first exposes a COOH-terminal glycine used in the conjugation of these proteins to lipids on the phagophore, the precursor to the autophagosome, whereas the second releases the lipid. The ATG4 family of proteases drives both cleavages, but how ATG4 proteins distinguish between soluble and lipid-anchored Atg8 proteins is not well understood. In a fully reconstituted delipidation assay, we establish that the physical anchoring of mammalian Atg8-family proteins in the membrane dramatically shifts the way ATG4 proteases recognize these substrates. Thus, while ATG4B is orders of magnitude faster at processing a soluble unprimed protein, all 4 ATG4 proteases can be activated to similar enzymatic activities on lipid-attached substrates. The recognition of lipidated but not soluble substrates is sensitive to a COOH-terminal LIR motif both in vitro and in cells. We suggest a model whereby ATG4B drives very fast priming of mammalian Atg8 proteins, whereas delipidation is inherently slow and regulated by all ATG4 homologs.
Collapse
Affiliation(s)
- Karlina J Kauffman
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA
| | - Shenliang Yu
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA
| | - Jiaxin Jin
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA.,b Lanzhou University Second Hospital , Lanzhou , Gansu Province , China
| | - Brian Mugo
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA
| | - Nathan Nguyen
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA
| | - Aidan O'Brien
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA
| | - Shanta Nag
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA
| | - Alf Håkon Lystad
- c Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo , Norway
| | - Thomas J Melia
- a Department of Cell Biology , Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|
37
|
Gao R, Ma J, Wen Z, Yang P, Zhao J, Xue M, Chen Y, Aldarouish M, Hu HM, Zhu XJ, Pan N, Wang LX. Tumor cell-released autophagosomes (TRAP) enhance apoptosis and immunosuppressive functions of neutrophils. Oncoimmunology 2018; 7:e1438108. [PMID: 29872581 DOI: 10.1080/2162402x.2018.1438108] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/28/2018] [Accepted: 02/02/2018] [Indexed: 01/22/2023] Open
Abstract
Our previous studies have confirmed that tumor cell-released autophagosomes (TRAP) could induce the differentiation of B cells into IL-10+ regulatory B cells (Bregs) with suppressive activities on T lymphocytes. However, the mechanism of TRAP-mediated immune suppression is still largely unclear. Herein, we sought to assess the immunomodulatory effect of TRAPs on human neutrophils, a major immune cell type that infiltrates human tumor tissues. We found that TRAPs enriched from malignant effusions or ascites of cancer patients and tumor cell lines were rapidly and effectively phagocytized by neutrophils through macropinocytosis and promoted neutrophil apoptosis via reactive oxygen species (ROS) generation and caspase-3 activation. Moreover, the apoptotic neutrophils that have phagocytized TRAPs inhibited the proliferation and activation of CD4+ T and CD8+ T cells in a cell contact- and ROS-dependent manner. These findings define a novel TRAP-mediated mechanism in neutrophils that potentially suppresses the anti-tumor T cell immunity and highlight TRAPs as an important target for future tumor immunotherapy.
Collapse
Affiliation(s)
- Rong Gao
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Jie Ma
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Zhifa Wen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Peiying Yang
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Jinjin Zhao
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Meng Xue
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Yongqiang Chen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Mohanad Aldarouish
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Hong-Ming Hu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China.,Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Xue-Jun Zhu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China.,Division of Hematology, Department of Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Ning Pan
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Li-Xin Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| |
Collapse
|
38
|
Autophagy induced during apoptosis degrades mitochondria and inhibits type I interferon secretion. Cell Death Differ 2017; 25:784-796. [PMID: 29229994 DOI: 10.1038/s41418-017-0017-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/10/2017] [Accepted: 10/18/2017] [Indexed: 02/08/2023] Open
Abstract
Cells undergoing Bax/Bak-mediated apoptosis exhibit signs of autophagy, but how it is activated and its significance is unknown. By directly activating Bax/Bak with BH3-only proteins or BH3 mimetic compounds, we demonstrate that mitochondrial damage correlated with a rapid increase in intracellular [AMP]/[ATP], phosphorylation of 5' AMP-activated protein kinase (AMPK), and activation of unc-51 like autophagy activating kinase 1 (ULK1). Consequently, autophagic flux was triggered early in the apoptotic pathway, as activation of the apoptosome and caspases were not necessary for its induction. Bax/Bak-triggered autophagy resulted in the clearance of damaged mitochondria in an ATG5/7-dependent manner that did not require Parkin. Importantly, Bax/Bak-mediated autophagy inhibited the secretion of the pro-inflammatory cytokine interferon-β (IFN-β) produced in response to mitochondrial damage, but not another cytokine interleukin-6 (IL-6). These findings show that Bax/Bak stimulated autophagy is essential for ensuring immunological silence during apoptosis.
Collapse
|
39
|
Vlahakis A, Lopez Muniozguren N, Powers T. Stress-response transcription factors Msn2 and Msn4 couple TORC2-Ypk1 signaling and mitochondrial respiration to ATG8 gene expression and autophagy. Autophagy 2017; 13:1804-1812. [PMID: 29198169 PMCID: PMC5788474 DOI: 10.1080/15548627.2017.1356949] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Macroautophagy/autophagy is a starvation and stress-induced catabolic process critical for cellular homeostasis and adaptation. Several Atg proteins are involved in the formation of the autophagosome and subsequent degradation of cytoplasmic components, a process termed autophagy flux. Additionally, the expression of several Atg proteins, in particular Atg8, is modulated transcriptionally, yet the regulatory mechanisms involved remain poorly understood. Here we demonstrate that the AGC kinase Ypk1, target of the rapamycin-insensitive TORC2 signaling pathway, controls ATG8 expression by repressing the heterodimeric Zinc-finger transcription factors Msn2 and Msn4. We find that Msn2 and Msn4 promote ATG8 expression downstream of the histone deacetylase complex (HDAC) subunit Ume6, a previously identified negative regulator of ATG8 expression. Moreover, we demonstrate that TORC2-Ypk1 signaling is functionally linked to distinct mitochondrial respiratory complexes. Surprisingly, we find that autophagy flux during amino acid starvation is also dependent upon Msn2-Msn4 activity, revealing a broad role for these transcription factors in the autophagy response.
Collapse
Affiliation(s)
- Ariadne Vlahakis
- a Department of Molecular and Cellular Biology , College of Biological Sciences, University of California, Davis , Davis , CA , USA.,b Department of Pathology , University of California, San Francisco , San Francisco , CA , USA
| | - Nerea Lopez Muniozguren
- a Department of Molecular and Cellular Biology , College of Biological Sciences, University of California, Davis , Davis , CA , USA
| | - Ted Powers
- a Department of Molecular and Cellular Biology , College of Biological Sciences, University of California, Davis , Davis , CA , USA
| |
Collapse
|
40
|
Abstract
In this issue of Molecular Cell, Torggler et al. (2016) leverage innovative synthetic biology approaches to dissect the spatiotemporal activation of Atg1 kinase during selective autophagy, revealing two distinct pathways that coordinately initiate autophagosome formation at the yeast vacuole.
Collapse
|
41
|
Postnikoff SD, Johnson JE, Tyler JK. The integrated stress response in budding yeast lifespan extension. MICROBIAL CELL (GRAZ, AUSTRIA) 2017; 4:368-375. [PMID: 29167799 PMCID: PMC5695854 DOI: 10.15698/mic2017.11.597] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/05/2017] [Indexed: 12/18/2022]
Abstract
Aging is a complex, multi-factorial biological process shared by all living organisms. It is manifested by a gradual accumulation of molecular alterations that lead to the decline of normal physiological functions in a time-dependent fashion. The ultimate goal of aging research is to develop therapeutic means to extend human lifespan, while reducing susceptibility to many age-related diseases including cancer, as well as metabolic, cardiovascular and neurodegenerative disorders. However, this first requires elucidation of the causes of aging, which has been greatly facilitated by the use of model organisms. In particular, the budding yeast Saccharomyces cerevisiae has been invaluable in the identification of conserved molecular and cellular determinants of aging and for the development of approaches to manipulate these aging determinants to extend lifespan. Strikingly, where examined, virtually all means to experimentally extend lifespan result in the induction of cellular stress responses. This review describes growing evidence in yeast that activation of the integrated stress response contributes significantly to lifespan extension. These findings demonstrate that yeast remains a powerful model system for elucidating conserved mechanisms to achieve lifespan extension that are likely to drive therapeutic approaches to extend human lifespan and healthspan.
Collapse
Affiliation(s)
- Spike D.L. Postnikoff
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065
| | - Jay E. Johnson
- Orentreich Foundation for the Advancement of Science, Cold Spring, NY
| | - Jessica K. Tyler
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065
| |
Collapse
|
42
|
Torggler R, Papinski D, Kraft C. Assays to Monitor Autophagy in Saccharomyces cerevisiae. Cells 2017; 6:cells6030023. [PMID: 28703742 PMCID: PMC5617969 DOI: 10.3390/cells6030023] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/04/2017] [Accepted: 07/09/2017] [Indexed: 12/18/2022] Open
Abstract
Autophagy is an intracellular process responsible for the degradation and recycling of cytoplasmic components. It selectively removes harmful cellular material and enables the cell to survive starvation by mobilizing nutrients via the bulk degradation of cytoplasmic components. While research over the last decades has led to the discovery of the key factors involved in autophagy, the pathway is not yet completely understood. The first studies of autophagy on a molecular level were conducted in the yeast Saccharomyces cerevisiae. Building up on these studies, many homologs have been found in higher eukaryotes. Yeast remains a highly relevant model organism for studying autophagy, with a wide range of established methods to elucidate the molecular details of the autophagy pathway. In this review, we provide an overview of methods to study both selective and bulk autophagy, including intermediate steps in the yeast Saccharomyces cerevisiae. We compare different assays, discuss their advantages and limitations and list potential applications.
Collapse
Affiliation(s)
- Raffaela Torggler
- Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Daniel Papinski
- Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria.
| | - Claudine Kraft
- Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria.
| |
Collapse
|
43
|
Noda T. Regulation of Autophagy through TORC1 and mTORC1. Biomolecules 2017; 7:biom7030052. [PMID: 28686223 PMCID: PMC5618233 DOI: 10.3390/biom7030052] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/01/2017] [Accepted: 07/04/2017] [Indexed: 12/27/2022] Open
Abstract
Autophagy is an intracellular protein-degradation process that is conserved across eukaryotes including yeast and humans. Under nutrient starvation conditions, intracellular proteins are transported to lysosomes and vacuoles via membranous structures known as autophagosomes, and are degraded. The various steps of autophagy are regulated by the target of rapamycin complex 1 (TORC1/mTORC1). In this review, a history of this regulation and recent advances in such regulation both in yeast and mammals will be discussed. Recently, the mechanism of autophagy initiation in yeast has been deduced. The autophagy-related gene 13 (Atg13) and the unc-51 like autophagy activating kinase 1 (Ulk1) are the most crucial substrates of TORC1 in autophagy, and by its dephosphorylation, autophagosome formation is initiated. Phosphorylation/dephosphorylation of Atg13 is regulated spatially inside the cell. Another TORC1-dependent regulation lies in the expression of autophagy genes and vacuolar/lysosomal hydrolases. Several transcriptional and post-transcriptional regulations are controlled by TORC1, which affects autophagy activity in yeast and mammals.
Collapse
Affiliation(s)
- Takeshi Noda
- Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
44
|
Yu S, Melia TJ. The coordination of membrane fission and fusion at the end of autophagosome maturation. Curr Opin Cell Biol 2017; 47:92-98. [PMID: 28463755 DOI: 10.1016/j.ceb.2017.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 02/16/2017] [Accepted: 03/21/2017] [Indexed: 10/19/2022]
Abstract
The two major objectives of macroautophagy are to sequester cargo away from the cytoplasm and deliver this material for breakdown in the lysosome. Sequestration is complete when the autophagosome membrane undergoes fission to produce separate inner and outer membranes, while delivery into the lysosome requires fusion of the outer autophagosome membrane with the lysosome membrane. Thus, the merging of membranes through fission and fusion underlies each of the pivotal events in macroautophagic clearance. How these merging events are controlled in the cell is poorly understood. Several recent studies however suggest that the two events may be temporally coordinated and rely upon members of the classic membrane fusion SNARE family as well as the autophagy-specific family of Atg8 proteins.
Collapse
Affiliation(s)
- Shenliang Yu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| | - Thomas J Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
45
|
Seo AY, Lau PW, Feliciano D, Sengupta P, Gros MAL, Cinquin B, Larabell CA, Lippincott-Schwartz J. AMPK and vacuole-associated Atg14p orchestrate μ-lipophagy for energy production and long-term survival under glucose starvation. eLife 2017; 6. [PMID: 28394250 PMCID: PMC5407857 DOI: 10.7554/elife.21690] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 04/09/2017] [Indexed: 12/22/2022] Open
Abstract
Dietary restriction increases the longevity of many organisms, but the cell signaling and organellar mechanisms underlying this capability are unclear. We demonstrate that to permit long-term survival in response to sudden glucose depletion, yeast cells activate lipid-droplet (LD) consumption through micro-lipophagy (µ-lipophagy), in which fat is metabolized as an alternative energy source. AMP-activated protein kinase (AMPK) activation triggered this pathway, which required Atg14p. More gradual glucose starvation, amino acid deprivation or rapamycin did not trigger µ-lipophagy and failed to provide the needed substitute energy source for long-term survival. During acute glucose restriction, activated AMPK was stabilized from degradation and interacted with Atg14p. This prompted Atg14p redistribution from ER exit sites onto liquid-ordered vacuole membrane domains, initiating µ-lipophagy. Our findings that activated AMPK and Atg14p are required to orchestrate µ-lipophagy for energy production in starved cells is relevant for studies on aging and evolutionary survival strategies of different organisms. DOI:http://dx.doi.org/10.7554/eLife.21690.001
Collapse
Affiliation(s)
- Arnold Y Seo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,Cell Biology and Metabolism Program, National Institutes of Health, Bethesda, United States
| | - Pick-Wei Lau
- Cell Biology and Metabolism Program, National Institutes of Health, Bethesda, United States
| | - Daniel Feliciano
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,Cell Biology and Metabolism Program, National Institutes of Health, Bethesda, United States
| | - Prabuddha Sengupta
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,Cell Biology and Metabolism Program, National Institutes of Health, Bethesda, United States
| | - Mark A Le Gros
- Department of Anatomy, University of California, San Francisco, San Francisco, United States.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| | - Bertrand Cinquin
- Department of Anatomy, University of California, San Francisco, San Francisco, United States
| | - Carolyn A Larabell
- Department of Anatomy, University of California, San Francisco, San Francisco, United States.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| | - Jennifer Lippincott-Schwartz
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,Cell Biology and Metabolism Program, National Institutes of Health, Bethesda, United States
| |
Collapse
|
46
|
Vlahakis A, Lopez Muniozguren N, Powers T. Calcium channel regulator Mid1 links TORC2-mediated changes in mitochondrial respiration to autophagy. J Cell Biol 2016; 215:779-788. [PMID: 27899413 PMCID: PMC5166500 DOI: 10.1083/jcb.201605030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/02/2016] [Accepted: 11/08/2016] [Indexed: 01/03/2023] Open
Abstract
Autophagy is a catabolic process that recycles cytoplasmic contents and is crucial for cell survival during stress. The target of rapamycin (TOR) kinase regulates autophagy as part of two distinct protein complexes, TORC1 and TORC2. TORC1 negatively regulates autophagy according to nitrogen availability. In contrast, TORC2 functions as a positive regulator of autophagy during amino acid starvation, via its target kinase Ypk1, by repressing the activity of the calcium-dependent phosphatase calcineurin and promoting the general amino acid control (GAAC) response. Precisely how TORC2-Ypk1 signaling regulates calcineurin within this pathway remains unknown. Here we demonstrate that activation of calcineurin requires Mid1, an endoplasmic reticulum-localized calcium channel regulatory protein implicated in the oxidative stress response. We find that normal mitochondrial respiration is perturbed in TORC2-Ypk1-deficient cells, which results in the accumulation of mitochondrial-derived reactive oxygen species that signal to Mid1 to activate calcineurin, thereby inhibiting the GAAC response and autophagy. These findings describe a novel pathway involving TORC2, mitochondrial oxidative stress, and calcium homeostasis for autophagy regulation.
Collapse
Affiliation(s)
- Ariadne Vlahakis
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, CA 95616
| | - Nerea Lopez Muniozguren
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, CA 95616
| | - Ted Powers
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, CA 95616
| |
Collapse
|
47
|
Koller B, Schramm C, Siebert S, Triebel J, Deland E, Pfefferkorn AM, Rickerts V, Thewes S. Dictyostelium discoideum as a Novel Host System to Study the Interaction between Phagocytes and Yeasts. Front Microbiol 2016; 7:1665. [PMID: 27818653 PMCID: PMC5073093 DOI: 10.3389/fmicb.2016.01665] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 10/05/2016] [Indexed: 01/26/2023] Open
Abstract
The social amoeba Dictyostelium discoideum is a well-established model organism to study the interaction between bacteria and phagocytes. In contrast, research using D. discoideum as a host model for fungi is rare. We describe a comprehensive study, which uses D. discoideum as a host model system to investigate the interaction with apathogenic (Saccharomyces cerevisiae) and pathogenic (Candida sp.) yeast. We show that Dictyostelium can be co-cultivated with yeasts on solid media, offering a convenient test to study the interaction between fungi and phagocytes. We demonstrate that a number of D. discoideum mutants increase (atg1-, kil1-, kil2-) or decrease (atg6-) the ability of the amoebae to predate yeast cells. On the yeast side, growth characteristics, reduced phagocytosis rate, as well as known virulence factors of C. albicans (EFG1, CPH1, HGC1, ICL1) contribute to the resistance of yeast cells against predation by the amoebae. Investigating haploid C. albicans strains, we suggest using the amoebae plate test for screening purposes after random mutagenesis. Finally, we discuss the potential of our adapted amoebae plate test to use D. discoideum for risk assessment of yeast strains.
Collapse
Affiliation(s)
- Barbara Koller
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität Berlin Berlin, Germany
| | - Christin Schramm
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität BerlinBerlin, Germany; FG16, Robert Koch InstituteBerlin, Germany
| | - Susann Siebert
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität Berlin Berlin, Germany
| | - János Triebel
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität Berlin Berlin, Germany
| | - Eric Deland
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität Berlin Berlin, Germany
| | - Anna M Pfefferkorn
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität Berlin Berlin, Germany
| | | | - Sascha Thewes
- Department of Biology, Chemistry, Pharmacy, Institute for Biology - Microbiology, Freie Universität Berlin Berlin, Germany
| |
Collapse
|
48
|
Dengjel J, Abeliovich H. Roles of mitophagy in cellular physiology and development. Cell Tissue Res 2016; 367:95-109. [PMID: 27488107 DOI: 10.1007/s00441-016-2472-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/04/2016] [Indexed: 12/19/2022]
Abstract
The autophagic degradation of mitochondria, or mitophagy, has been shown to occur in eukaryotic cells under various physiological conditions. Broadly, these fall into two categories: quality-control related mitophagy and developmentally induced mitophagy. Quality-control related mitophagy, which is the lysosomal/vacuolar degradation of malfunctioning or superfluous mitochondria, is an important housekeeping function in respiring eukaryotic cells. It plays an essential role in physiological homeostasis and its deregulation has been linked to the progression of late-onset diseases. On the other hand, developmental processes such as reticulocyte maturation have also been shown to involve mitophagy. Importantly, there are clear differences between these processes. Unlike our knowledge of the more general degradation of soluble cytosolic content during starvation-induced macroautophagy, the mechanisms involved in the selective autophagic degradation of mitochondria have only recently begun to receive significant attention. Here, we review the current literature on these topics and proceed to provide specific examples from yeast and mammalian systems. Finally, we cover experimental approaches, with a focus on proteomic methods dedicated to the study of mitophagy in different systems.
Collapse
Affiliation(s)
- Jörn Dengjel
- Department of Dermatology, Medical Center, Freiburg Institute for Advanced Studies (FRIAS), ZBSA Center for Biological Systems Analysis, University of Freiburg, Habsburgerstr. 49, 79104, Freiburg, Germany.,Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Hagai Abeliovich
- Department of Biochemistry and Food Science, Hebrew University of Jerusalem, Rehovot, 76100, Israel. .,FRIAS Senior Fellow and Marie Curie Fellow of the European Union Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Albertstr. 19, 79104, Freiburg, Germany.
| |
Collapse
|
49
|
Yang S, Rosenwald AG. Autophagy in Saccharomyces cerevisiae requires the monomeric GTP-binding proteins, Arl1 and Ypt6. Autophagy 2016; 12:1721-1737. [PMID: 27462928 PMCID: PMC5079543 DOI: 10.1080/15548627.2016.1196316] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Macroautophagy/autophagy is a cellular degradation process that sequesters organelles or proteins into a double-membrane structure called the phagophore; this transient compartment matures into an autophagosome, which then fuses with the lysosome or vacuole to allow hydrolysis of the cargo. Factors that control membrane traffic are also essential for each step of autophagy. Here we demonstrate that 2 monomeric GTP-binding proteins in Saccharomyces cerevisiae, Arl1 and Ypt6, which belong to the Arf/Arl/Sar protein family and the Rab family, respectively, and control endosome-trans-Golgi traffic, are also necessary for starvation-induced autophagy under high temperature stress. Using established autophagy-specific assays we found that cells lacking either ARL1 or YPT6, which exhibit synthetic lethality with one another, were unable to undergo autophagy at an elevated temperature, although autophagy proceeds normally at normal growth temperature; specifically, strains lacking one or the other of these genes are unable to construct the autophagosome because these 2 proteins are required for proper traffic of Atg9 to the phagophore assembly site (PAS) at the restrictive temperature. Using degron technology to construct an inducible arl1Δ ypt6Δ double mutant, we demonstrated that cells lacking both genes show defects in starvation-inducted autophagy at the permissive temperature. We also found Arl1 and Ypt6 participate in autophagy by targeting the Golgi-associated retrograde protein (GARP) complex to the PAS to regulate the anterograde trafficking of Atg9. Our data show that these 2 membrane traffic regulators have novel roles in autophagy.
Collapse
Affiliation(s)
- Shu Yang
- a Department of Biology , Georgetown University , Washington DC , USA
| | - Anne G Rosenwald
- a Department of Biology , Georgetown University , Washington DC , USA
| |
Collapse
|
50
|
Shi Q, Song X, Liu Z, Wang Y, Wang Y, Fu J, Su C, Xia X, Song E, Song Y. Quinones Derived from Polychlorinated Biphenyls Induce ROS-Dependent Autophagy by Evoking an Autophagic Flux and Inhibition of mTOR/p70S6k. Chem Res Toxicol 2016; 29:1160-71. [DOI: 10.1021/acs.chemrestox.6b00127] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Qiong Shi
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Xiufang Song
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Zixuan Liu
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Yawen Wang
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Yuxin Wang
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Juanli Fu
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Chuanyang Su
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Xiaomin Xia
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Erqun Song
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| | - Yang Song
- Key Laboratory
of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People’s Republic of China
| |
Collapse
|