Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Autophagy ; 20(7): 1471-1472, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38744436

ABSTRACT

The destination of a damaged lysosome is either being repaired if the damage is small or degraded through a lysosome-specific macroautophagy/autophagy pathway named lysophagy when the damage is too extensive to repair. Even though previous studies report lumenal glycan exposure during lysosome damage as a signal to trigger lysophagy, it is possibly beneficial for cells to initiate lysophagy earlier than membrane rupture. In a recently published article, Gahlot et al. determined that SPART/SPG20 senses lipid-packing defects and recruits and activates the ubiquitin ligase ITCH, which labels damaged lysosomes with ubiquitin chains to initiate lysophagy.


Subject(s)
Autophagy , Lysosomes , Lysosomes/metabolism , Humans , Autophagy/physiology , Animals , Macroautophagy/physiology , Ubiquitin-Protein Ligases/metabolism , Models, Biological , Ubiquitin/metabolism
2.
Autophagy ; 20(7): 1559-1576, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38522078

ABSTRACT

A large proportion of patients with chronic pain experience co-morbid anxiety. The medial prefrontal cortex (mPFC) is proposed to underlie this comorbidity, but the molecular and neuronal mechanisms are not fully understood. Here, we reported that impaired neuronal macroautophagy in the prelimbic cortical (PrL) subregion of the mPFC paralleled the occurrence of anxiety-like behaviors in rats with chronic spared nerve injury (SNI). Intriguingly, such macroautophagy impairment was mainly observed in a FOS/c-Fos+ neuronal subpopulation in the PrL. Chemogenetic inactivation of this comorbid anxiety-related neuronal ensemble relieved pain-induced anxiety-like behaviors. Rescuing macroautophagy impairment in this neuronal ensemble relieved chronic pain-associated anxiety and mechanical allodynia and restored synaptic homeostasis at the molecular level. By contrast, artificial disruption of macroautophagy induced early-onset co-morbid anxiety in neuropathic rats, but not general anxiety in normal rats. Taken together, our work identifies causal linkage between PrL neuronal macroautophagy dysfunction and comorbid anxiety in neuropathic pain and provides novel insights into the role of PrL by differentiating its contribution in pain-induced comorbid anxiety from its modulation over general anxiety-like behaviors.Abbreviation: AAV: adeno-associated viruses; ACC: anterior cingulate cortex; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG12: autophagy related 12; CAMK2/CaMKII: calcium/calmodulin-dependent protein kinase II; CNO: clozapine-N-oxide; CQ: chloroquine; DIA: data independent acquisition; DIO: double floxed inverse orf; DLG4/PSD-95: discs large MAGUK scaffold protein 4; Dox: doxycycline; GABA: γ-aminobutyric acid; GFP: green fluorescent protein; GO: gene ontology; Gi: inhibitory guanine nucleotide-binding proteins; HsCHRM4/M4D: human cholinergic receptor muscarinic 4; HsSYN: human synapsin; KEGG: Kyoto encyclopedia of genes and genomes; LAMP1: lysosomal-associated membrane protein 1; LC3-II: PE conjugated microtubule-associated protein 1 light chain3; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; mPFC: medial prefrontal cortex; P2A: 2A self-cleaving peptide; PPI: protein-protein interaction networks; PrL: prelimbic cortex; RBFOX3/NeuN: RNA binding protein, fox-1 homolog (C. elegans) 3; rtTA: reverse tetracycline-transactivator; SDS-PAGE: sodium dodecylsulfate-polyacrylamide gel electrophoresis; SHANK3: SH3 and multiple ankyrin repeat domains 3; SLC1A1/EAAC1: solute carrier family 1 (neuronal/epithelial high affinity glutamate transporter, systemXag), member 1; SNAP23: synaptosomal-associated protein 23; SNI:spared nerve injury; SQSTM1/p62: sequestosome 1; SYT3: synaptotagmin 3; TRE: tetracycline-responsive element; TRE3G: third-generation tetracycline-responsive element.


Subject(s)
Anxiety , Macroautophagy , Neuralgia , Neurons , Prefrontal Cortex , Animals , Neuralgia/metabolism , Prefrontal Cortex/metabolism , Rats , Neurons/metabolism , Male , Macroautophagy/physiology , Rats, Sprague-Dawley , Behavior, Animal , Chronic Pain/metabolism , Autophagy/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hyperalgesia
3.
Autophagy ; 20(6): 1457-1458, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38348842

ABSTRACT

The lysosomal degradation of the endoplasmic reticulum (ER), known as "reticulophagy", is important for protein quality control and organelle turnover. Here we present a noncanonical reticulophagy occurring at ER exit sites (ERESs) induced by the misfolded SERPINA1/α1-antitrypsin (AAT) mutant, Z-AAT. The accumulation of Z-AAT arrests ER-to-Golgi transport, and recruits V-ATPase and ATG16L1 to mediate LC3C decoration of ERESs. Consequently, the receptor RETREG1/FAM134B-2 is recruited by lipidated LC3C to initiate reticulophagy. Furthermore, the blockade of ER export acts as a universal signal to activate reticulophagy mediated by the V-ATPase-ATG16L1-LC3C axis. This study sheds light on the mechanism of how ERESs switch from ER export to reticulophagy for quality control.


Subject(s)
Autophagy-Related Proteins , Endoplasmic Reticulum , Microtubule-Associated Proteins , Vacuolar Proton-Translocating ATPases , Endoplasmic Reticulum/metabolism , Humans , Autophagy-Related Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Golgi Apparatus/metabolism , Autophagy/physiology , alpha 1-Antitrypsin/metabolism , Animals , Membrane Proteins/metabolism , Lysosomes/metabolism , Macroautophagy/physiology , Signal Transduction , Intracellular Signaling Peptides and Proteins
4.
Autophagy ; 20(5): 1197-1198, 2024 May.
Article in English | MEDLINE | ID: mdl-38163952

ABSTRACT

Under stress conditions, the endoplasmic reticulum and nucleus undergo turnover through selective macroautophagy/autophagy processes termed reticulophagy and nucleophagy, respectively. Our recent study has identified the protein Hva22/Rop1/Yep1, a member of the REEP1-REEP4 subfamily of the REEP protein family, as an essential factor for both processes in the fission yeast Schizosaccharomyces pombe. In the absence of Hva22/Yep1, reticulophagy and nucleophagy cargos without surrounding autophagic membranes accumulate in the cytoplasm. Interestingly, human proteins in the REEP1-REEP4 subfamily can functionally substitute for Hva22/Yep1 to facilitate reticulophagy. Phylogenetic and synteny analyses further reveal that the budding yeast reticulophagy receptor Atg40 is also a REEP1-REEP4 subfamily member. Similar to human REEP1-REEP4 subfamily proteins, Atg40 can functionally replace Hva22/Yep1. Based on our findings, we propose that promoting reticulophagy is a conserved function of REEP1-REEP4 subfamily proteins.


Subject(s)
Autophagy , Schizosaccharomyces , Schizosaccharomyces/metabolism , Humans , Autophagy/physiology , Schizosaccharomyces pombe Proteins/metabolism , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Macroautophagy/physiology
5.
Autophagy ; 20(5): 1208-1209, 2024 May.
Article in English | MEDLINE | ID: mdl-38293799

ABSTRACT

Selective degradation of the endoplasmic reticulum (ER) by macroautophagy/autophagy (reticulophagy) is essential for maintaining ER morphology and homeostasis under environmental stresses. Several reticulophagy receptors have been identified in mammals and yeast, but their counterparts in plants have not been extensively explored yet. Recently, we demonstrated that the HVA22-family protein OsHLP1 is a reticulophagy receptor in rice plants, and its orthologs function similarly in Arabidopsis plants. In this punctum, we discuss why the HVA22 family proteins are the reticulophagy receptors in plants and how reticulophagy is highly associated with plant immune response.


Subject(s)
Endoplasmic Reticulum , Endoplasmic Reticulum/metabolism , Autophagy/physiology , Plant Proteins/metabolism , Macroautophagy/physiology , Arabidopsis/metabolism , Arabidopsis/genetics , Plants/metabolism , Animals
6.
Proc Natl Acad Sci U S A ; 119(26): e2111506119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35737835

ABSTRACT

Macroautophagy promotes cellular homeostasis by delivering cytoplasmic constituents to lysosomes for degradation [Mizushima, Nat. Cell Biol. 20, 521-527 (2018)]. However, while most studies have focused on the mechanisms of protein degradation during this process, we report here that macroautophagy also depends on glycan degradation via the glycosidase, α-l-fucosidase 1 (FUCA1), which removes fucose from glycans. We show that cells lacking FUCA1 accumulate lysosomal glycans, which is associated with impaired autophagic flux. Moreover, in a mouse model of fucosidosis-a disease characterized by inactivating mutations in FUCA1 [Stepien et al., Genes (Basel) 11, E1383 (2020)]-glycan and autophagosome/autolysosome accumulation accompanies tissue destruction. Mechanistically, using lectin capture and mass spectrometry, we identified several lysosomal enzymes with altered fucosylation in FUCA1-null cells. Moreover, we show that the activity of some of these enzymes in the absence of FUCA1 can no longer be induced upon autophagy stimulation, causing retardation of autophagic flux, which involves impaired autophagosome-lysosome fusion. These findings therefore show that dysregulated glycan degradation leads to defective autophagy, which is likely a contributing factor in the etiology of fucosidosis.


Subject(s)
Fucosidosis , Macroautophagy , Polysaccharides , Animals , Fucosidosis/genetics , Fucosidosis/metabolism , Lysosomes/metabolism , Macroautophagy/physiology , Mice , Polysaccharides/metabolism , alpha-L-Fucosidase/genetics , alpha-L-Fucosidase/metabolism
7.
Biochem Biophys Res Commun ; 614: 161-168, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35597153

ABSTRACT

Vacuoles and lysosomes are organelles involved in the degradation of cytoplasmic proteins and organelles. Vacuolar morphology is dynamically regulated by fission and fusion in budding yeast. Vacuolar fusion is elicited in nutrient-depleted conditions and mediated by inactivation of target of rapamycin complex 1 (TORC1) protein kinase. However, it is unknown whether and how vacuolar morphology affects macroautophagy and microautophagy, which are induced by nutrient starvation and TORC1 inactivation. Here, we developed a system to control vacuolar fission in budding yeast. Vacuolar fragmentation promoted microautophagy but not macroautophagy. Vacuolar fragmentation caused multiple nucleus-vacuole junctions. Multiple vacuoles caused by vacuolar fragmentation also improved micronucleophagy (microautophagic degradation of a portion of the nucleus). However, vacuolar morphology did not impact nucleolar remodeling, condensation of the rDNA (rRNA gene) region, or separation of ribosomal DNA from nucleolar proteins, which is evoked by TORC1 inactivation. Thus, this study provides insights into the impacts of vacuolar/lysosomal morphology on macroautophagy and microautophagy.


Subject(s)
Macroautophagy , Microautophagy , Saccharomyces cerevisiae Proteins , Saccharomycetales , Vacuoles , Autophagy , DNA, Ribosomal/genetics , Macroautophagy/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Microautophagy/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomycetales/metabolism , Vacuoles/metabolism
8.
Biochem Pharmacol ; 197: 114899, 2022 03.
Article in English | MEDLINE | ID: mdl-34968496

ABSTRACT

The accumulation of aggregated α-synuclein (α-syn) has been identified as the primary component of Lewy bodies that are the pathological hallmarks of Parkinson's disease (PD). Several preclinical studies have shown α-syn aggregation, and particularly the intermediates formed during the aggregation process to be toxic to cells. Current PD treatments only provide symptomatic relief, and α-syn serves as a promising target to develop a disease-modifying therapy for PD. Our previous studies have revealed that a small-molecular inhibitor for prolyl oligopeptidase (PREP), KYP-2047, increases α-syn degradation by accelerating macroautophagy (MA) leading to disease-modifying effects in preclinical PD models. However, α-syn is also degraded by chaperone-mediated autophagy (CMA). In the present study, we tested the effects of PREP inhibition or deletion on CMA activation and α-syn degradation. HEK-293 cells were transfected with α-syn and incubated with 1 & 10 µM KYP-2047 for 24 h. Both 1 & 10 µM KYP-2047 increased LAMP-2A levels, induced α-syn degradation and reduced the expression of Hsc70, suggesting that the PREP inhibitor prevented α-syn aggregation by activating the CMA pathway. Similarly, KYP-2047 increased the LAMP-2A immunoreactivity and reduced the Hsc70 levels in mouse primary cortical neurons. When LAMP-2A was silenced by a siRNA, KYP-2047 increased the LC3BII/LC3BI ratio and accelerated the clearance of α-syn. Additionally, KYP-2047 induced CMA effectively also when MA was blocked by bafilomycin A1. Based on our results, we suggest that PREP might function as a core network node in MA-CMA crosstalk, and PREP inhibition can reduce α-syn levels via both main autophagy systems.


Subject(s)
Chaperone-Mediated Autophagy/physiology , Macroautophagy/physiology , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Cells, Cultured , Chaperone-Mediated Autophagy/drug effects , Gene Knockout Techniques , HEK293 Cells , Humans , Macroautophagy/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Proline/analogs & derivatives , Proline/pharmacology
9.
Food Chem Toxicol ; 158: 112706, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34848256

ABSTRACT

α-Synuclein, which is associated with Parkinson's disease, is cleared by the ubiquitin-proteasome system and autophagy lysosome system. Chaperon-mediated autophagy (CMA) and macroautophagy are major subtypes of autophagy and play a critical role in pesticide-induced α-synucleinopathy. In this study, we explored the role of CMA in diquat (DQ)-induced α-synucleinopathy and characterized the relationship between CMA and macroautophagy in the clearance of pathologic α-synuclein for the prevention of DQ neurotoxicity. DQ was cytotoxic to SH-SY5Y cells in a concentration-dependent manner, as shown by decreased cell viability and increased cytotoxicity. DQ treatment was also found to induce autophagy such as CMA and macroautophagy by monitoring the expression of Lamp2A and microtubule-associated protein 1A/1B light chain 3B (LC3-II) respectively. Following DQ treatment, SH-SY5Y cells were found to have induced phosphorylated and detergent-insoluble α-synuclein deposits, and MG132, a proteasome inhibitor, effectively potentiated both CMA and macroautophagy for preventing α-synuclein aggregation. Interestingly, CMA impairment by Lamp2A-knock down decreased the LC3II expression compared to in DQ-treated cells transfected with control siRNA. In Lamp2-knock down cells, pathologic α-synuclein was increased 12 h after DQ treatment, but there was no change observed at 24 h. In DQ-treated cells, macroautophagy by 3-methyladenine and bafilomycin inhibition increased Lamp2A expression, indicating an increase in CMA activity. In addition, CMA modulation affected apoptosis, and inhibiting lysosome activity by NH4Cl increased apoptosis in DQ-treated cells. An increase in autophagy was confirmed to compensate for the decrease in lysosome activity. Pretreatment with z-VAD-fmk, a pan-caspase inhibitor, significantly enhanced the macroautophagy response of DQ-exposed cells without alterations in Lamp2A expression. Our results suggest that CMA can regulate DQ-induced α-synucleinopathy cooperatively with macroautophagy, and crosstalk between macroautophagy and CMA plays an important role in DQ-induced cytotoxicity. Taken together, autophagy modulation may be a useful treatment strategy in pesticide-induced neurodegenerative disorders through preventing α-synucleinopathy.


Subject(s)
Apoptosis/drug effects , Chaperone-Mediated Autophagy , Diquat/toxicity , Macroautophagy , alpha-Synuclein , Cell Line, Tumor , Cell Survival/drug effects , Chaperone-Mediated Autophagy/drug effects , Chaperone-Mediated Autophagy/physiology , Humans , Macroautophagy/drug effects , Macroautophagy/physiology , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/metabolism
10.
Elife ; 102021 09 29.
Article in English | MEDLINE | ID: mdl-34585663

ABSTRACT

Removal of damaged organelles via the process of selective autophagy constitutes a major form of cellular quality control. Damaged organelles are recognized by a dedicated surveillance machinery, leading to the assembly of an autophagosome around the damaged organelle, prior to fusion with the degradative lysosomal compartment. Lysosomes themselves are also prone to damage and are degraded through the process of lysophagy. While early steps involve recognition of ruptured lysosomal membranes by glycan-binding galectins and ubiquitylation of transmembrane lysosomal proteins, many steps in the process, and their interrelationships, remain poorly understood, including the role and identity of cargo receptors required for completion of lysophagy. Here, we employ quantitative organelle capture and proximity biotinylation proteomics of autophagy adaptors, cargo receptors, and galectins in response to acute lysosomal damage, thereby revealing the landscape of lysosome-associated proteome remodeling during lysophagy. Among the proteins dynamically recruited to damaged lysosomes were ubiquitin-binding autophagic cargo receptors. Using newly developed lysophagic flux reporters including Lyso-Keima, we demonstrate that TAX1BP1, together with its associated kinase TBK1, are both necessary and sufficient to promote lysophagic flux in both HeLa cells and induced neurons (iNeurons). While the related receptor Optineurin (OPTN) can drive damage-dependent lysophagy when overexpressed, cells lacking either OPTN or CALCOCO2 still maintain significant lysophagic flux in HeLa cells. Mechanistically, TAX1BP1-driven lysophagy requires its N-terminal SKICH domain, which binds both TBK1 and the autophagy regulatory factor RB1CC1, and requires upstream ubiquitylation events for efficient recruitment and lysophagic flux. These results identify TAX1BP1 as a central component in the lysophagy pathway and provide a proteomic resource for future studies of the lysophagy process.


Subject(s)
Autophagy/genetics , Lysosomes/pathology , Macroautophagy/physiology , Ubiquitin/metabolism , Humans , Protein Binding , Proteomics
11.
Autophagy ; 17(8): 2048-2050, 2021 08.
Article in English | MEDLINE | ID: mdl-34074213

ABSTRACT

TMEM41B and VMP1, two endoplasmic reticulum (ER)-resident transmembrane proteins, play important roles in regulating the formation of lipid droplets (LDs), autophagy initiation, and viral infection. However, the biochemical functions of TMEM41B and VMP1 are unclear. A lipids distribution screen suggested TMEM41B and VMP1 are critical to the normal distribution of cholesterol and phosphatidylserine. Biochemical analyses unveiled that TMEM41B and VMP1 have scramblase activity. These findings shed light on the mechanism by which TMEM41B and VMP1 regulate LD formation, lipids distribution, macroautophagy, and viral infection.


Subject(s)
Autophagy/physiology , Membrane Proteins/metabolism , Phospholipid Transfer Proteins/metabolism , Animals , Autophagosomes/metabolism , Humans , Macroautophagy/physiology
12.
Biochem Biophys Res Commun ; 545: 69-74, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33545634

ABSTRACT

Peroxisomes play an essential role in cellular homeostasis by regulating lipid metabolism and the conversion of reactive oxygen species (ROS). Several peroxisomal proteins, known as peroxins (PEXs), control peroxisome biogenesis and degradation. Various mutations in the PEX genes are genetic causes for the development of inheritable peroxisomal-biogenesis disorders, such as Zellweger syndrome. Among the peroxins, PEX1 defects are the most common mutations in Zellweger syndrome. PEX1 is an AAA-ATPase that regulates the recycling of PEX5, which is essential for importing peroxisome matrix proteins. However, the post-transcriptional regulation of PEX1 is largely unknown. Here, we showed that heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) controls PEX1 expression. In addition, we found that depletion of HNRNPA1 induces autophagic degradation of peroxisome, which is blocked in ATG5-knockout cells. In addition, depletion of HNRNPA1 increased peroxisomal ROS levels. Inhibition of the generation of peroxisomal ROS by treatment with NAC significantly suppressed pexophagy in HNRNPA1-deficient cells. Taken together, our results suggest that depletion of HNRNPA1 increases peroxisomal ROS and pexophagy by downregulating PEX1 expression.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Macroautophagy/physiology , Membrane Proteins/metabolism , Peroxisomes/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Cells, Cultured , Down-Regulation , Gene Knockout Techniques , HCT116 Cells , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein A1/deficiency , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Humans , Macroautophagy/genetics , Membrane Proteins/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Zellweger Syndrome/genetics , Zellweger Syndrome/metabolism
13.
Autophagy ; 17(11): 3461-3474, 2021 11.
Article in English | MEDLINE | ID: mdl-33509017

ABSTRACT

Macroautophagy/autophagy plays an important role in the control of viral infections and viruses have evolved multiple strategies to interfere with autophagy to avoid destruction and promote their own replication and spread. Here we report that the deubiquitinase encoded in the N-terminal domain of the Epstein-Barr virus (EBV) large tegument protein, BPLF1, regulates selective autophagy. Mass spectrometry analysis identified several vesicular traffic and autophagy related proteins as BPLF1 interactors and potential substrates, suggesting that the viral protein targets this cellular defense during productive infection. Direct binding of BPLF1 to the autophagy receptor SQSTM1/p62 (sequestosome 1) was confirmed by co-immunoprecipitation of transfected BPLF1 and by in vitro affinity isolation of bacterially expressed proteins. Expression of the catalytically active BPLF1 was associated with decreased SQSTM1/p62 ubiquitination and failure to recruit LC3 to SQSTM1/p62-positive aggregates. Selective autophagy was inhibited as illustrated by the accumulation of large protein aggregates in BPLF1-positive cells co-transfected with an aggregate-prone HTT (huntingtin)-Q109 construct, and by a slower autophagy-dependent clearance of protein aggregates upon transfection of BPLF1 in cells expressing a tetracycline-regulated HTT-Q103. The inhibition of aggregate clearance was restored by overexpression of a SQSTM1/p62[E409A,K420R] mutant that does not require ubiquitination of Lys420 for cargo loading. These findings highlight a previously unrecognized role of the viral deubiquitinase in the regulation of selective autophagy, which may promote infection and the production of infectious virus.Abbreviations: BPLF1, BamH1 fragment left open reading frame-1; EBV, Epstein-Barr virus; GFP, green fluorescent protein; HTT, huntingtin; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; PB1, Phox and Bem1 domain; PE, phosphatidylethanolamine; SQSTM1/p62, sequestosome 1; UBA, ubiquitin-associated domain.


Subject(s)
Autophagy/physiology , Deubiquitinating Enzymes/physiology , Herpesvirus 4, Human/physiology , Sequestosome-1 Protein/physiology , Viral Regulatory and Accessory Proteins/physiology , Autophagy/genetics , Deubiquitinating Enzymes/genetics , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , HeLa Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Macroautophagy/genetics , Macroautophagy/physiology , Microtubule-Associated Proteins/metabolism , Mutation , Protein Aggregates/genetics , Protein Aggregates/physiology , Sequestosome-1 Protein/genetics , Transfection , Ubiquitination , Viral Regulatory and Accessory Proteins/genetics
14.
Nat Commun ; 12(1): 183, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420039

ABSTRACT

We have reported that autophagy is crucial for clearance of amyloidogenic human IAPP (hIAPP) oligomer, suggesting that an autophagy enhancer could be a therapeutic modality against human diabetes with amyloid accumulation. Here, we show that a recently identified autophagy enhancer (MSL-7) reduces hIAPP oligomer accumulation in human induced pluripotent stem cell-derived ß-cells (hiPSC-ß-cells) and diminishes oligomer-mediated apoptosis of ß-cells. Protective effects of MSL-7 against hIAPP oligomer accumulation and hIAPP oligomer-mediated ß-cell death are significantly reduced in cells with knockout of MiTF/TFE family members such as Tfeb or Tfe3. MSL-7 improves glucose tolerance and ß-cell function of hIAPP+ mice on high-fat diet, accompanied by reduced hIAPP oligomer/amyloid accumulation and ß-cell apoptosis. Protective effects of MSL-7 against hIAPP oligomer-mediated ß-cell death and the development of diabetes are also significantly reduced by ß-cell-specific knockout of Tfeb. These results suggest that an autophagy enhancer could have therapeutic potential against human diabetes characterized by islet amyloid accumulation.


Subject(s)
Amyloid/metabolism , Amyloidogenic Proteins/metabolism , Autophagy/physiology , Diabetes Mellitus, Type 2/metabolism , Islet Amyloid Polypeptide/genetics , Islet Amyloid Polypeptide/metabolism , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Gene Knockout Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells , Macroautophagy/physiology , Mice , Mice, Transgenic
15.
Theranostics ; 11(1): 222-256, 2021.
Article in English | MEDLINE | ID: mdl-33391472

ABSTRACT

Macroautophagy (hereafter called autophagy) is a highly conserved physiological process that degrades over-abundant or damaged organelles, large protein aggregates and invading pathogens via the lysosomal system (the vacuole in plants and yeast). Autophagy is generally induced by stress, such as oxygen-, energy- or amino acid-deprivation, irradiation, drugs, etc. In addition to non-selective bulk degradation, autophagy also occurs in a selective manner, recycling specific organelles, such as mitochondria, peroxisomes, ribosomes, endoplasmic reticulum (ER), lysosomes, nuclei, proteasomes and lipid droplets (LDs). This capability makes selective autophagy a major process in maintaining cellular homeostasis. The dysfunction of selective autophagy is implicated in neurodegenerative diseases (NDDs), tumorigenesis, metabolic disorders, heart failure, etc. Considering the importance of selective autophagy in cell biology, we systemically review the recent advances in our understanding of this process and its regulatory mechanisms. We emphasize the 'cargo-ligand-receptor' model in selective autophagy for specific organelles or cellular components in yeast and mammals, with a focus on mitophagy and ER-phagy, which are finely described as types of selective autophagy. Additionally, we highlight unanswered questions in the field, helping readers focus on the research blind spots that need to be broken.


Subject(s)
Macroautophagy/physiology , Mitophagy/physiology , Autophagy/physiology , Humans , Organelles
17.
Autophagy ; 17(5): 1157-1169, 2021 05.
Article in English | MEDLINE | ID: mdl-32264736

ABSTRACT

The fusion of autophagosomes and endosomes/lysosomes, also called autophagosome maturation, ensures the degradation of autophagic cargoes. It is an important regulatory step of the macroautophagy/autophagy process. STX17 is the key autophagosomal SNARE protein that mediates autophagosome maturation. Here, we report that the acetylation of STX17 regulates its SNARE activity and autophagic degradation. The histone acetyltransferase CREBBP/CBP and the deacetylase HDAC2 specifically regulate the acetylation of STX17. In response to cell starvation and MTORC1 inhibition, the inactivation of CREBBP leads to the deacetylation of STX17 at its SNARE domain. This deacetylation promotes the interaction between STX17 and SNAP29 and the formation of the STX17-SNAP29-VAMP8 SNARE complex with no effect on the recruitment of STX17 to autophagosomal membranes. Deacetylation of STX17 also enhances the interaction between STX17 and the tethering complex HOPS, thereby further promoting autophagosome-lysosome fusion. Our study suggests a mechanism by which acetylation regulates the late-stage of autophagy, and possibly other STX17-related intracellular membrane fusion events.Abbreviations: ACTB: actin beta; CREBBP/CBP: CREB binding protein; Ctrl: control; GFP: green fluorescent protein; GST: glutathione S-transferase; HDAC: histone deacetylase; HOPS: homotypic fusion and protein sorting complex; KO: knockout; LAMP1/2: lysosomal associated membrane protein 1/2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEFs: mouse embryonic fibroblasts; MS: mass spectrometry; MTORC1: mechanistic target of rapamycin kinase complex 1; NAM: nicotinamide; PtdIns3K: phosphatidylinositol 3-kinase; RFP: red fluorescent protein; SNAP29: synaptosome associated protein 29; SNARE: soluble N-ethylamide-sensitive factor attachment protein receptor; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TSA: trichostatin A; TSC1/2: TSC complex subunit 1/2; VAMP8: vesicle associated membrane protein 8; WT: wild type.


Subject(s)
Autophagosomes/metabolism , Autophagy/physiology , Macroautophagy/physiology , Qa-SNARE Proteins/metabolism , Endosomes/metabolism , Fibroblasts/metabolism , Humans , Lysosomes/metabolism , Membrane Fusion/physiology
18.
Drug Discov Today ; 26(3): 704-712, 2021 03.
Article in English | MEDLINE | ID: mdl-33301978

ABSTRACT

Growing multidrug-resistant (MDR) strains of various infectious bacterial species are hindering research aiming to eliminate such infections. During a bacterial infection, the host response eliminates the pathogen via fusion of the endocytic vesicles with lysosomes, called xenophagy. However, MDR bacteria have evolved strategies to escape xenophagy. In this review, we propose novel therapeutics for overcoming such escape, including chimeric antibiotics, nanoformulations for the induction of autophagy in infected cells, and small interfering (si)RNA-mediated silencing of genes to inhibit the host-pathogen interaction. We also discuss the role of combinations of antibiotics showing synergy, the administrative routes of differentially capped nanoparticles (NPs), and the use of different types of nanoformulations for eliminating pathogenic bacteria from the host.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacterial Infections/drug therapy , Anti-Bacterial Agents/administration & dosage , Autophagy/drug effects , Bacteria/pathogenicity , Bacterial Infections/microbiology , Drug Resistance, Multiple, Bacterial , Drug Synergism , Drug Therapy, Combination , Host-Pathogen Interactions , Humans , Lysosomes , Macroautophagy/physiology , Nanoparticles
19.
Autophagy ; 17(11): 3297-3305, 2021 11.
Article in English | MEDLINE | ID: mdl-33228439

ABSTRACT

Selective autophagy is a specific elimination of certain intracellular substrates by autophagic pathways. The most studied macroautophagy pathway involves tagging and recognition of a specific cargo by the autophagic membrane (phagophore) followed by the complete sequestration of targeted cargo from the cytosol by the double-membrane vesicle, autophagosome. Until recently, the knowledge about selective macroautophagy was minimal, but now there is a panoply of links elucidating how phagophores engulf their substrates selectively. The studies of selective autophagy processes have further stressed the importance of using the in vivo models to validate new in vitro findings and discover the physiologically relevant mechanisms. However, dissecting how the selective autophagy occurs yet remains difficult in living organisms, because most of the organelles are relatively inaccessible to observation and experimental manipulation in mammals. In recent years, zebrafish (Danio rerio) is widely recognized as an excellent model for studying autophagic processes in vivo because of its optical accessibility, genetic manipulability and translational potential. Several selective autophagy pathways, such as mitophagy, xenophagy, lipophagy and aggrephagy, have been investigated using zebrafish and still need to be studied further, while other selective autophagy pathways, such as pexophagy or reticulophagy, could also benefit from the use of the zebrafish model. In this review, we shed light on how zebrafish contributed to our understanding of these selective autophagy processes by providing the in vivo platform to study them at the organismal level and highlighted the versatility of zebrafish model in the selective autophagy field.Abbreviations: AD: Alzheimer disease; ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CMA: chaperone-mediated autophagy; CQ: chloroquine; HsAMBRA1: human AMBRA1; KD: knockdown; KO: knockout; LD: lipid droplet; MMA: methylmalonic acidemia; PD: Parkinson disease; Tg: transgenic.


Subject(s)
Autophagy/physiology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Autophagy/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Lipid Droplets/physiology , Macroautophagy/genetics , Macroautophagy/physiology , Mitophagy/genetics , Mitophagy/physiology , Models, Animal , Models, Biological , Protein Aggregates/genetics , Protein Aggregates/physiology , Zebrafish/genetics
20.
Autophagy ; 16(11): 2102-2104, 2020 11.
Article in English | MEDLINE | ID: mdl-32718210

ABSTRACT

Macroautophagy/autophagy is an intracellular degradative pathway that is often induced as a pro-survival process for cells under stress. A few recent reports establish the role of the glycogen metabolic pathway in neuronal cell survival in conditions such as oxidative stress and hypoxia, and the possible link between glycogen synthesis and autophagy induction. This commentary highlights the emerging role of GYS (glycogen synthase) in neuronal autophagy and stress response.


Subject(s)
Autophagy/physiology , Glycogen/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Animals , Cell Survival/physiology , Humans , Macroautophagy/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...