Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Nat Rev Mol Cell Biol ; 21(8): 439-458, 2020 08.
Article in English | MEDLINE | ID: mdl-32372019

ABSTRACT

Autophagosomes are double-membrane vesicles newly formed during autophagy to engulf a wide range of intracellular material and transport this autophagic cargo to lysosomes (or vacuoles in yeasts and plants) for subsequent degradation. Autophagosome biogenesis responds to a plethora of signals and involves unique and dynamic membrane processes. Autophagy is an important cellular mechanism allowing the cell to meet various demands, and its disruption compromises homeostasis and leads to various diseases, including metabolic disorders, neurodegeneration and cancer. Thus, not surprisingly, the elucidation of the molecular mechanisms governing autophagosome biogenesis has attracted considerable interest. Key molecules and organelles involved in autophagosome biogenesis, including autophagy-related (ATG) proteins and the endoplasmic reticulum, have been discovered, and their roles and relationships have been investigated intensely. However, several fundamental questions, such as what supplies membranes/lipids to build the autophagosome and how the membrane nucleates, expands, bends into a spherical shape and finally closes, have proven difficult to address. Nonetheless, owing to recent studies with new approaches and technologies, we have begun to unveil the mechanisms underlying these processes on a molecular level. We now know that autophagosome biogenesis is a highly complex process, in which multiple proteins and lipids from various membrane sources, supported by the formation of membrane contact sites, cooperate with biophysical phenomena, including membrane shaping and liquid-liquid phase separation, to ensure seamless segregation of the autophagic cargo. Together, these studies pave the way to obtaining a holistic view of autophagosome biogenesis.


Subject(s)
Autophagosomes/metabolism , Autophagosomes/physiology , Animals , Autophagy , Autophagy-Related Proteins/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Humans , Lysosomes/metabolism , Macroautophagy , Protein Transport
2.
Cell ; 167(6): 1433-1435, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912049

ABSTRACT

This year's Nobel Prize in Physiology or Medicine has been awarded to Yoshinori Ohsumi for the discovery of the molecular principles governing autophagy, an intracellular degradation pathway routed via lysosomes or vacuoles. It is a story of a simple yet insightful yeast genetic screen that revealed the inner circuitry of one of the most powerful quality-control pathways in cells.


Subject(s)
Autophagy , Nobel Prize , Physiology/history , Animals , Autophagosomes/physiology , History, 20th Century , Humans , Lysosomes/physiology , Yeasts/cytology , Yeasts/physiology
3.
EMBO J ; 40(15): e108050, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34155657

ABSTRACT

Selective autophagy mediates specific degradation of unwanted cytoplasmic components to maintain cellular homeostasis. The suppressor of gene silencing 3 (SGS3) and RNA-dependent RNA polymerase 6 (RDR6)-formed bodies (SGS3/RDR6 bodies) are essential for siRNA amplification in planta. However, whether autophagy receptors regulate selective turnover of SGS3/RDR6 bodies is unknown. By analyzing the transcriptomic response to virus infection in Arabidopsis, we identified a virus-induced small peptide 1 (VISP1) composed of 71 amino acids, which harbor a ubiquitin-interacting motif that mediates interaction with autophagy-related protein 8. Overexpression of VISP1 induced selective autophagy and compromised antiviral immunity by inhibiting SGS3/RDR6-dependent viral siRNA amplification, whereas visp1 mutants exhibited opposite effects. Biochemistry assays demonstrate that VISP1 interacted with SGS3 and mediated autophagic degradation of SGS3/RDR6 bodies. Further analyses revealed that overexpression of VISP1, mimicking the sgs3 mutant, impaired biogenesis of endogenous trans-acting siRNAs and up-regulated their targets. Collectively, we propose that VISP1 is a small peptide receptor functioning in the crosstalk between selective autophagy and RNA silencing.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Arabidopsis/immunology , Peptides/genetics , RNA-Dependent RNA Polymerase/metabolism , Arabidopsis/metabolism , Arabidopsis/virology , Arabidopsis Proteins/genetics , Autophagosomes/physiology , Autophagy/physiology , Autophagy-Related Protein 8 Family/metabolism , Gene Expression Regulation, Plant , Mutation , Peptides/metabolism , Plant Immunity , Plants, Genetically Modified , RNA, Small Interfering , RNA-Dependent RNA Polymerase/genetics , Nicotiana/genetics
4.
EMBO J ; 40(4): e105120, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33368531

ABSTRACT

Autophagy is a lysosome-dependent degradation pathway essential to maintain cellular homeostasis. Therefore, either defective or excessive autophagy may be detrimental for cells and tissues. The past decade was characterized by significant advances in molecular dissection of stimulatory autophagy inputs; however, our understanding of the mechanisms that restrain autophagy is far from complete. Here, we describe a negative feedback mechanism that limits autophagosome biogenesis based on the selective autophagy-mediated degradation of ATG13, a component of the ULK1 autophagy initiation complex. We demonstrate that the centrosomal protein OFD1 acts as bona fide autophagy receptor for ATG13 via direct interaction with the Atg8/LC3/GABARAP family of proteins. We also show that patients with Oral-Facial-Digital type I syndrome, caused by mutations in the OFD1 gene, display excessive autophagy and that genetic inhibition of autophagy in a mouse model of the disease, significantly ameliorates polycystic kidney, a clinical manifestation of the disorder. Collectively, our data report the discovery of an autophagy self-regulated mechanism and implicate dysregulated autophagy in the pathogenesis of renal cystic disease in mammals.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagosomes/physiology , Autophagy-Related Protein 8 Family/metabolism , Autophagy , Microtubule-Associated Proteins/metabolism , Polycystic Kidney Diseases/pathology , Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Autophagy-Related Protein 8 Family/genetics , Humans , Lysosomes/metabolism , Lysosomes/pathology , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Polycystic Kidney Diseases/etiology , Polycystic Kidney Diseases/metabolism , Proteins/genetics
5.
Proc Natl Acad Sci U S A ; 119(30): e2201927119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858426

ABSTRACT

Hepatitis B virus (HBV) DNA replication takes place inside the viral core particle and is dependent on autophagy. Here we show that HBV core particles are associated with autophagosomes and phagophores in cells that productively replicate HBV. These autophagic membrane-associated core particles contain almost entirely the hypophosphorylated core protein and are DNA replication competent. As the hyperphosphorylated core protein can be localized to phagophores and the dephosphorylation of the core protein is associated with the packaging of viral pregenomic RNA (pgRNA), these results are in support of the model that phagophores can serve as the sites for the packaging of pgRNA. In contrast, in cells that replicate HBV, the precore protein derivatives, which are related to the core protein, are associated with autophagosomes but not with phagophores via a pathway that is independent of its signal peptide. Interestingly, when the core protein is expressed by itself, it is associated with phagophores but not with autophagosomes. These observations indicate that autophagic membranes are differentially involved in the trafficking of precore and core proteins. HBV induces the fusion of autophagosomes and multivesicular bodies and the silencing of Rab11, a regulator of this fusion, is associated with the reduction of release of mature HBV particles. Our studies thus indicate that autophagic membranes participate in the assembly of HBV nucleocapsids, the trafficking of HBV precore and core proteins, and likely also the egress of HBV particles.


Subject(s)
Autophagosomes , Hepatitis B virus , Nucleocapsid , Viral Genome Packaging , Virus Replication , Autophagosomes/physiology , DNA, Viral/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , Nucleocapsid/genetics , Nucleocapsid/physiology , Protein Transport , RNA, Viral/metabolism , Virus Replication/genetics
6.
Trends Biochem Sci ; 45(6): 484-496, 2020 06.
Article in English | MEDLINE | ID: mdl-32307224

ABSTRACT

Autophagy is traditionally depicted as a signaling cascade that culminates in the formation of an autophagosome that degrades cellular cargo. However, recent studies have identified myriad pathways and cellular organelles underlying the autophagy process, be it as signaling platforms or through the contribution of proteins and lipids. The Golgi complex is recognized as being a central transport hub in the cell, with a critical role in endocytic trafficking and endoplasmic reticulum (ER) to plasma membrane (PM) transport. However, the Golgi is also an important site of key autophagy regulators, including the protein autophagy-related (ATG)-9A and the lipid, phosphatidylinositol-4-phosphate [PI(4)P]. In this review, we highlight the central function of this organelle in autophagy as a transport hub supplying various components of autophagosome formation.


Subject(s)
Autophagosomes/physiology , Golgi Apparatus/physiology , Autophagy , Autophagy-Related Proteins/physiology , Biological Transport , Endosomes/metabolism , Humans , Lipid Metabolism , Membrane Proteins/physiology , Vesicular Transport Proteins/physiology
7.
Proc Natl Acad Sci U S A ; 117(35): 21391-21402, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817423

ABSTRACT

Syntaxin17, a key autophagosomal N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein, can associate with ATG8 family proteins SNAP29 and VAMP8 to facilitate the membrane fusion process between the double-membraned autophagosome and single-membraned lysosome in mammalian macroautophagy. However, the inherent properties of Syntaxin17 and the mechanistic basis underlying the interactions of Syntaxin17 with its binding proteins remain largely unknown. Here, using biochemical, NMR, and structural approaches, we systemically characterized Syntaxin17 as well as its interactions with ATG8 family proteins, SNAP29 and VAMP8. We discovered that Syntaxin17 alone adopts an autoinhibited conformation mediated by a direct interaction between its Habc domain and the Qa-SNARE motif. In addition, we revealed that the Qa-SNARE region of Syntaxin17 contains one LC3-interacting region (LIR) motif, which preferentially binds to GABARAP subfamily members. Importantly, the GABARAP binding of Syntaxin17 can release its autoinhibited state. The determined crystal structure of the Syntaxin17 LIR-GABARAP complex not only provides mechanistic insights into the interaction between Syntaxin17 and GABARAP but also reveals an unconventional LIR motif with a C-terminally extended 310 helix for selectively binding to ATG8 family proteins. Finally, we also elucidated structural arrangements of the autophagic Syntaxin17-SNAP29-VAMP8 SNARE core complex, and uncovered its conserved biochemical and structural characteristics common to all other SNAREs. In all, our findings reveal three distinct states of Syntaxin17, and provide mechanistic insights into the Syntaxin17-mediated autophagosome-lysosome fusion process.


Subject(s)
Autophagosomes/physiology , Lysosomes/physiology , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , R-SNARE Proteins/metabolism , Amino Acid Motifs , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 8 Family/metabolism , Escherichia coli , Humans , Microtubule-Associated Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 117(26): 15230-15241, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32513711

ABSTRACT

Mutations in UBQLN2 cause amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and other neurodegenerations. However, the mechanism by which the UBQLN2 mutations cause disease remains unclear. Alterations in proteins involved in autophagy are prominent in neuronal tissue of human ALS UBQLN2 patients and in a transgenic P497S UBQLN2 mouse model of ALS/FTD, suggesting a pathogenic link. Here, we show UBQLN2 functions in autophagy and that ALS/FTD mutant proteins compromise this function. Inactivation of UBQLN2 expression in HeLa cells reduced autophagic flux and autophagosome acidification. The defect in acidification was rescued by reexpression of wild type (WT) UBQLN2 but not by any of the five different UBQLN2 ALS/FTD mutants tested. Proteomic analysis and immunoblot studies revealed P497S mutant mice and UBQLN2 knockout HeLa and NSC34 cells have reduced expression of ATP6v1g1, a critical subunit of the vacuolar ATPase (V-ATPase) pump. Knockout of UBQLN2 expression in HeLa cells decreased turnover of ATP6v1g1, while overexpression of WT UBQLN2 increased biogenesis of ATP6v1g1 compared with P497S mutant UBQLN2 protein. In vitro interaction studies showed that ATP6v1g1 binds more strongly to WT UBQLN2 than to ALS/FTD mutant UBQLN2 proteins. Intriguingly, overexpression of ATP6v1g1 in UBQLN2 knockout HeLa cells increased autophagosome acidification, suggesting a therapeutic approach to overcome the acidification defect. Taken together, our findings suggest that UBQLN2 mutations drive pathogenesis through a dominant-negative loss-of-function mechanism in autophagy and that UBQLN2 functions as an important regulator of the expression and stability of ATP6v1g1. These findings may have important implications for devising therapies to treat UBQLN2-linked ALS/FTD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyotrophic Lateral Sclerosis/genetics , Autophagosomes/physiology , Autophagy-Related Proteins/metabolism , Autophagy/genetics , Dementia/genetics , Adaptor Proteins, Signal Transducing/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy-Related Proteins/genetics , Biomarkers/metabolism , Cell Line , Dementia/metabolism , Dementia/pathology , Genetic Predisposition to Disease , Humans , Hydrogen-Ion Concentration , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Protein Binding , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Up-Regulation , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
9.
Plant J ; 105(4): 1083-1097, 2021 02.
Article in English | MEDLINE | ID: mdl-33222335

ABSTRACT

Plant responses to NH4+ stress are complex, and multiple mechanisms underlying NH4+ sensitivity and tolerance in plants may be involved. Here, we demonstrate that macro- and microautophagic activities are oppositely affected in plants grown under NH4+ toxicity conditions. When grown under NH4+ stress conditions, macroautophagic activity was impaired in roots. Root cells accumulated autophagosomes in the cytoplasm, but showed less autophagic flux, indicating that late steps of the macroautophagy process are affected under NH4+ stress conditions. Under this scenario, we also found that the CCZ1-MON1 complex, a critical factor for vacuole delivery pathways, functions in the late step of the macroautophagic pathway in Arabidopsis. In contrast, an accumulation of tonoplast-derived vesicles was observed in vacuolar lumens of root cells of NH4+ -stressed plants, suggesting the induction of a microautophagy-like process. In this sense, some SYP22-, but mainly VAMP711-positive vesicles were observed inside vacuole in roots of NH4+ -stressed plants. Consistent with the increased tonoplast degradation and the reduced membrane flow to the vacuole due to the impaired macroautophagic flux, the vacuoles of root cells of NH4+ -stressed plants showed a simplified structure and lower tonoplast content. Taken together, this study presents evidence that postulates late steps of the macroautophagic process as a relevant physiological mechanism underlying the NH4+ sensitivity response in Arabidopsis, and additionally provides insights into the molecular tools for studying microautophagy in plants.


Subject(s)
Ammonium Compounds/metabolism , Arabidopsis/metabolism , Microautophagy , Plant Roots/metabolism , Arabidopsis/physiology , Autophagosomes/metabolism , Autophagosomes/physiology , Plant Roots/physiology , Stress, Physiological
10.
Mol Biol Evol ; 38(1): 128-141, 2021 01 04.
Article in English | MEDLINE | ID: mdl-32722765

ABSTRACT

Spermatogenesis is an essential process for producing sperm cells. Reproductive strategy is successfully evolved for a species to adapt to a certain ecological system. However, roles of newly evolved genes in testis autophagy remain unclear. In this study, we found that a newly evolved gene srag (Sox9-regulated autophagy gene) plays an important role in promoting autophagy in testis in the lineage of the teleost Monopterus albus. The gene integrated into an interaction network through a two-way strategy of evolution, via Sox9-binding in its promoter and interaction with Becn1 in the coding region. Its promoter region evolved a cis element for binding of Sox9, a transcription factor for male sex determination. Both in vitro and in vivo analyses demonstrated that transcription factor Sox9 could bind to and activate the srag promoter. Its coding region acquired ability to interact with key autophagy initiation factor Becn1 via the conserved C-terminal, indicating that srag integrated into preexisting autophagy network. Moreover, we determined that Srag enhanced autophagy by interacting with Becn1. Notably, srag transgenic zebrafish revealed that Srag exerted the same function by enhancing autophagy through the Srag-Becn1 pathway. Thus, the new gene srag regulated autophagy in testis by integrated into preexisting autophagy network.


Subject(s)
Autophagy/genetics , Biological Evolution , Eels/physiology , SOX9 Transcription Factor/metabolism , Testis/physiology , Animals , Animals, Genetically Modified , Autophagosomes/physiology , Male , Zebrafish
11.
EMBO J ; 37(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29343546

ABSTRACT

The removal of misfolded, ubiquitinated proteins is an essential part of the protein quality control. The ubiquitin-proteasome system (UPS) and autophagy are two interconnected pathways that mediate the degradation of such proteins. During autophagy, ubiquitinated proteins are clustered in a p62-dependent manner and are subsequently engulfed by autophagosomes. However, the nature of the protein substrates targeted for autophagy is unclear. Here, we developed a reconstituted system using purified components and show that p62 and ubiquitinated proteins spontaneously coalesce into larger clusters. Efficient cluster formation requires substrates modified with at least two ubiquitin chains longer than three moieties and is based on p62 filaments cross-linked by the substrates. The reaction is inhibited by free ubiquitin, K48-, and K63-linked ubiquitin chains, as well as by the autophagosomal marker LC3B, suggesting a tight cross talk with general proteostasis and autophagosome formation. Our study provides mechanistic insights on how substrates are channeled into autophagy.


Subject(s)
Autophagy/physiology , Protein Aggregation, Pathological/prevention & control , RNA-Binding Proteins/metabolism , Ubiquitinated Proteins/metabolism , Autophagosomes/physiology , Cell Line, Tumor , Humans , Microtubule-Associated Proteins/metabolism , Protein Aggregation, Pathological/pathology , Protein Folding , Ubiquitin/metabolism
12.
EMBO J ; 37(7)2018 04 03.
Article in English | MEDLINE | ID: mdl-29467218

ABSTRACT

In 1900, Adami speculated that a sequence of context-independent energetic and structural changes governed the reversion of differentiated cells to a proliferative, regenerative state. Accordingly, we show here that differentiated cells in diverse organs become proliferative via a shared program. Metaplasia-inducing injury caused both gastric chief and pancreatic acinar cells to decrease mTORC1 activity and massively upregulate lysosomes/autophagosomes; then increase damage associated metaplastic genes such as Sox9; and finally reactivate mTORC1 and re-enter the cell cycle. Blocking mTORC1 permitted autophagy and metaplastic gene induction but blocked cell cycle re-entry at S-phase. In kidney and liver regeneration and in human gastric metaplasia, mTORC1 also correlated with proliferation. In lysosome-defective Gnptab-/- mice, both metaplasia-associated gene expression changes and mTORC1-mediated proliferation were deficient in pancreas and stomach. Our findings indicate differentiated cells become proliferative using a sequential program with intervening checkpoints: (i) differentiated cell structure degradation; (ii) metaplasia- or progenitor-associated gene induction; (iii) cell cycle re-entry. We propose this program, which we term "paligenosis", is a fundamental process, like apoptosis, available to differentiated cells to fuel regeneration following injury.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Regeneration/physiology , Acinar Cells , Animals , Autophagosomes/physiology , Cell Cycle/physiology , Cell Transdifferentiation/physiology , Cellular Reprogramming/physiology , Chief Cells, Gastric/pathology , Gastrointestinal Tract/pathology , Gene Expression , Humans , Lysosomes , Metaplasia/genetics , Mice , Mice, Inbred C57BL , S Phase/physiology , SOX9 Transcription Factor/metabolism , Stomach/injuries , Stomach/pathology , Transferases (Other Substituted Phosphate Groups)/genetics
13.
J Neurosci ; 40(42): 8103-8118, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32917789

ABSTRACT

Interstitial axon branching is an essential step during the establishment of neuronal connectivity. However, the exact mechanisms on how the number and position of branches are determined are still not fully understood. Here, we investigated the role of Arl8B, an adaptor molecule between lysosomes and kinesins. In chick retinal ganglion cells (RGCs), downregulation of Arl8B reduces axon branch density and shifts their location more proximally, while Arl8B overexpression leads to increased density and more distal positions of branches. These alterations correlate with changes in the location and density of lysosomes and autophagosomes along the axon shaft. Diminishing autophagy directly by knock-down of atg7, a key autophagy gene, reduces branch density, while induction of autophagy by rapamycin increases axon branching, indicating that autophagy plays a prominent role in axon branch formation. In vivo, local inactivation of autophagy in the retina using a mouse conditional knock-out approach disturbs retino-collicular map formation which is dependent on the formation of interstitial axon branches. These data suggest that Arl8B plays a principal role in the positioning of axon branches by spatially controlling autophagy, thus directly controlling formation of neural connectivity in the brain.SIGNIFICANCE STATEMENT The formation of interstitial axonal branches plays a prominent role in numerous places of the developing brain during neural circuit establishment. We show here that the GTPase Arl8B controls density and location of interstitial axon branches, and at the same time controls also density and location of the autophagy machinery. Upregulation or downregulation of autophagy in vitro promotes or inhibits axon branching. Local disruption of autophagy in vivo disturbs retino-collicular mapping. Our data suggest that Arl8B controls axon branching by controlling locally autophagy. This work is one of the first reports showing a role of autophagy during early neural circuit development and suggests that autophagy in general plays a much more prominent role during brain development than previously anticipated.


Subject(s)
ADP-Ribosylation Factors/physiology , Autophagosomes/physiology , Axons/physiology , Lysosomes/physiology , ADP-Ribosylation Factors/metabolism , Animals , Autophagosomes/enzymology , Autophagosomes/ultrastructure , Autophagy/genetics , Axons/enzymology , Axons/ultrastructure , Chick Embryo , Down-Regulation , Gene Knockdown Techniques , Lysosomes/enzymology , Lysosomes/ultrastructure , Mice, Knockout , Primary Cell Culture , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/ultrastructure
14.
J Neurochem ; 157(3): 752-763, 2021 05.
Article in English | MEDLINE | ID: mdl-33354770

ABSTRACT

Fused in sarcoma (FUS) is a ubiquitously expressed RNA/DNA-binding protein that plays different roles in the cell. FUS pathology has been reported in neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Mutations in FUS have also been linked to a subset of familial ALS. FUS is mainly localized in the nucleus although it shuttles between the nucleus and the cytoplasm. ALS-linked mutations cause the accumulation of the FUS protein in cytoplasm where it forms stress granule-like inclusions. The protein- and RNA-containing inclusions are reported to be positive of autophagosome markers and degraded by the autophagy pathway. However, the role of FUS in the autophagy pathway remains to be better understood. Using immunoblot and confocal imaging techniques in this study, we found that FUS knockout (KO) cells showed a decreased basal autophagy level. Rapamycin and bafilomycin A1 treatment showed that FUS KO cells were not able to initiate autophagy as efficiently as wild-type cells, suggesting that the autophagosome formation is affected in the absence of FUS. Moreover, using immunoblot and quantitative PCR techniques, we found that the mRNA and protein levels of the genes critical in the initial steps of the autophagy pathway (FIP200, ATG16L1 and ATG12) were significantly lower in FUS KO cells. Re-expressing FUS in the KO cells restored the expression of FIP200 and ATG16L1. Our findings demonstrate a novel role of FUS in the autophagy pathway, that is, regulating the transcription of genes involved in early stages of autophagy such as the initiation and elongation of autophagosomes.


Subject(s)
Autophagosomes/genetics , Autophagosomes/physiology , Autophagy/genetics , Autophagy/physiology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/physiology , Animals , Autophagosomes/drug effects , Autophagy/drug effects , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/physiology , Cell Line , Gene Expression Regulation , Gene Knockout Techniques , Macrolides/pharmacology , Mice , Proteasome Endopeptidase Complex , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction/genetics , Sirolimus/pharmacology
15.
Cancer Sci ; 112(10): 3972-3978, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34407274

ABSTRACT

While starvation-induced autophagy is thought to randomly degrade cellular components, under certain circumstances autophagy selectively recognizes, sequesters, and degrades specific targets via autophagosomes. This process is called selective autophagy, and it contributes to cellular homeostasis by degrading specific soluble proteins, supramolecular complexes, liquid-liquid phase-separated droplets, abnormal or excess organelles, and pathogenic invasive bacteria. This means that autophagy, like the ubiquitin-proteasome system, strictly regulates diverse cellular functions through its selectivity. In this short review, we focus on the mechanism of "selective" autophagy, which is rapidly being elucidated.


Subject(s)
Autophagosomes/physiology , Autophagy/physiology , Autophagy-Related Protein 8 Family/metabolism , Cell Physiological Phenomena , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Homeostasis/physiology , Humans , Organelles , Phagocytosis/physiology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ubiquitination
16.
Eur J Immunol ; 50(8): 1174-1186, 2020 08.
Article in English | MEDLINE | ID: mdl-32250445

ABSTRACT

Staphylococcus aureus, a pathogen most frequently found in diabetic foot ulcer infection, was recently suggested as an intracellular pathogen. Autophagy in professional phagocytes like macrophages allows selective destruction of intracellular pathogens, and its dysfunction can increase the survival of internalized pathogens, causing infections to worsen and spread. Previous works have shown that S. aureus infections in diabetes appeared more severe and invasive, and coincided with the suppressed autophagy in dermal tissues of diabetic rat, but the exact mechanisms are unclear. Here, we demonstrated that accumulation of advanced glycation end products (AGEs) contributed to the diminished autophagy-mediated clearance of S. aureus in the macrophages differentiated from PMA-treated human monocytic cell line THP-1. Importantly, infected macrophages showed increased S. aureus containing autophagosome, but the subsequent fusion of S. aureus containing autophagosome and lysosome was suppressed in AGEs-pretreated cells, suggesting AGEs blocked the autophagic flux and enabled S. aureus survival and escape. At the molecular level, elevated lysosomal ARL8 expression in AGEs-treated macrophages was required for AGEs-mediated inhibition of autophagosome-lysosome fusion. Silencing ARL8 in AGEs-treated macrophages restored autophagic flux and increased S. aureus clearance. Our results therefore demonstrate a new mechanism, in which AGEs accelerate S. aureus immune evasion in macrophages by ARL8-dependent suppression of autophagosome-lysosome fusion and bactericidal capability.


Subject(s)
ADP-Ribosylation Factors/physiology , Glycation End Products, Advanced/physiology , Lysosomes/physiology , Macrophages/immunology , Phagocytosis , Staphylococcus aureus/immunology , Autophagosomes/physiology , Humans , Immune Evasion , THP-1 Cells , Up-Regulation
17.
FASEB J ; 34(1): 161-179, 2020 01.
Article in English | MEDLINE | ID: mdl-31914609

ABSTRACT

Fas-apoptotic inhibitory molecule 2 (FAIM2) is a member of the transmembrane BAX inhibitor motif-containing (TMBIM) family. TMBIM family is comprised of six anti-apoptotic proteins that suppress cell death by regulating endoplasmic reticulum Ca2+ homeostasis. Recent studies have implicated two TMBIM proteins, GRINA and BAX Inhibitor-1, in mediating cytoprotection via autophagy. However, whether FAIM2 plays a role in autophagy has been unknown. Here we show that FAIM2 localizes to the lysosomes at basal state and facilitates autophagy through interaction with microtubule-associated protein 1 light chain 3 proteins in human neuroblastoma SH-SY5Y cells. FAIM2 overexpression increased autophagy flux, while autophagy flux was impaired in shRNA-mediated knockdown (shFAIM2) cells, and the impairment was more evident in the presence of rapamycin. In shFAIM2 cells, autophagosome maturation through fusion with lysosomes was impaired, leading to accumulation of autophagosomes. A functional LC3-interacting region motif within FAIM2 was essential for the interaction with LC3 and rescue of autophagy flux in shFAIM2 cells while LC3-binding property of FAIM2 was dispensable for the anti-apoptotic function in response to Fas receptor-mediated apoptosis. Suppression of autophagosome maturation was also observed in a null mutant of Caenorhabditis elegans lacking xbx-6, the ortholog of FAIM2. Our study suggests that FAIM2 is a novel regulator of autophagy mediating autophagosome maturation through the interaction with LC3.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagosomes/physiology , Lysosomes/physiology , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Amino Acid Motifs , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Caenorhabditis elegans/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Immunosuppressive Agents/pharmacology , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Plasmids , Protein Transport , Sirolimus/pharmacology
18.
Brain ; 143(3): 783-799, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32185393

ABSTRACT

Frontotemporal dementia and amyotrophic lateral sclerosis are clinically and pathologically overlapping disorders with shared genetic causes. We previously identified a disease locus on chromosome 16p12.1-q12.2 with genome-wide significant linkage in a large European Australian family with autosomal dominant inheritance of frontotemporal dementia and amyotrophic lateral sclerosis and no mutation in known amyotrophic lateral sclerosis or dementia genes. Here we demonstrate the segregation of a novel missense variant in CYLD (c.2155A>G, p.M719V) within the linkage region as the genetic cause of disease in this family. Immunohistochemical analysis of brain tissue from two CYLD p.M719V mutation carriers showed widespread glial CYLD immunoreactivity. Primary mouse neurons transfected with CYLDM719V exhibited increased cytoplasmic localization of TDP-43 and shortened axons. CYLD encodes a lysine 63 deubiquitinase and CYLD cutaneous syndrome, a skin tumour disorder, is caused by mutations that lead to reduced deubiquitinase activity. In contrast with CYLD cutaneous syndrome-causative mutations, CYLDM719V exhibited significantly increased lysine 63 deubiquitinase activity relative to the wild-type enzyme (paired Wilcoxon signed-rank test P = 0.005). Overexpression of CYLDM719V in HEK293 cells led to more potent inhibition of the cell signalling molecule NF-κB and impairment of autophagosome fusion to lysosomes, a key process in autophagy. Although CYLD mutations appear to be rare, CYLD's interaction with at least three other proteins encoded by frontotemporal dementia and/or amyotrophic lateral sclerosis genes (TBK1, OPTN and SQSTM1) suggests that it may play a central role in the pathogenesis of these disorders. Mutations in several frontotemporal dementia and amyotrophic lateral sclerosis genes, including TBK1, OPTN and SQSTM1, result in a loss of autophagy function. We show here that increased CYLD activity also reduces autophagy function, highlighting the importance of autophagy regulation in the pathogenesis of frontotemporal dementia and amyotrophic lateral sclerosis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Deubiquitinating Enzyme CYLD/genetics , Deubiquitinating Enzyme CYLD/physiology , Frontotemporal Dementia/genetics , Genetic Predisposition to Disease/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Autophagosomes/metabolism , Autophagosomes/physiology , Axons/pathology , Brain/metabolism , DNA-Binding Proteins , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzymes/metabolism , Frontotemporal Dementia/metabolism , Mice , Mutation, Missense/genetics , NF-kappa B/antagonists & inhibitors , Primary Cell Culture , Transfection
19.
PLoS Genet ; 14(4): e1007359, 2018 04.
Article in English | MEDLINE | ID: mdl-29694367

ABSTRACT

The autophagosomal SNARE Syntaxin17 (Syx17) forms a complex with Snap29 and Vamp7/8 to promote autophagosome-lysosome fusion via multiple interactions with the tethering complex HOPS. Here we demonstrate that, unexpectedly, one more SNARE (Ykt6) is also required for autophagosome clearance in Drosophila. We find that loss of Ykt6 leads to large-scale accumulation of autophagosomes that are unable to fuse with lysosomes to form autolysosomes. Of note, loss of Syx5, the partner of Ykt6 in ER-Golgi trafficking does not prevent autolysosome formation, pointing to a more direct role of Ykt6 in fusion. Indeed, Ykt6 localizes to lysosomes and autolysosomes, and forms a SNARE complex with Syx17 and Snap29. Interestingly, Ykt6 can be outcompeted from this SNARE complex by Vamp7, and we demonstrate that overexpression of Vamp7 rescues the fusion defect of ykt6 loss of function cells. Finally, a point mutant form with an RQ amino acid change in the zero ionic layer of Ykt6 protein that is thought to be important for fusion-competent SNARE complex assembly retains normal autophagic activity and restores full viability in mutant animals, unlike palmitoylation or farnesylation site mutant Ykt6 forms. As Ykt6 and Vamp7 are both required for autophagosome-lysosome fusion and are mutually exclusive subunits in a Syx17-Snap29 complex, these data suggest that Vamp7 is directly involved in membrane fusion and Ykt6 acts as a non-conventional, regulatory SNARE in this process.


Subject(s)
Autophagosomes/physiology , Drosophila Proteins/physiology , Lysosomes/physiology , Membrane Fusion/physiology , R-SNARE Proteins/physiology , Animals , Animals, Genetically Modified , Binding Sites , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Membrane Fusion/genetics , Models, Biological , Multiprotein Complexes/genetics , Multiprotein Complexes/physiology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/physiology , R-SNARE Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SNARE Proteins/genetics , SNARE Proteins/physiology
20.
J Cell Physiol ; 235(10): 6767-6778, 2020 10.
Article in English | MEDLINE | ID: mdl-31985039

ABSTRACT

Ferroptosis has been reported as a unique form of cell death. However, in recent years, researchers have increasingly challenged the uniqueness of ferroptosis compared to other types of cell death. In this study, we examined whether ferroptosis shares cell death pathways with other types of cell death, especially autophagy, via the autophagic process. Here, we observed that ferroptosis inducers (artesunate [ART] and erastin [ERA]) and autophagy inducers (bortezomib [BOR] and XIE62-1004) led to autophagosome formation via the endoplasmic reticulum (ER) stress response. Unlike XIE62-1004, ART, ERA, and BOR, which affect glutathione production or utilization, induced oxidative stress responses-an increase in the levels of heme oxygenase-1 and lipid peroxidation. Oxidative stress responses were attenuated by deletion of autophagy-related gene-5 or treatment with autophagy inhibitors (bafilomycin and chloroquine). Our studies provide an overview of common death pathways-the ER stress response-associated autophagic process in ferroptosis and autophagy. We also highlight the role played by glutathione redox system in the outcome of the autophagic process.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum Stress/physiology , Ferroptosis/physiology , Apoptosis/physiology , Autophagosomes/metabolism , Autophagosomes/physiology , Cell Line, Tumor , Glutathione/metabolism , HCT116 Cells , Heme Oxygenase-1/metabolism , Humans , Lipid Peroxidation/physiology , Oxidation-Reduction , Oxidative Stress/physiology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL