Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Microb Pathog ; 190: 106638, 2024 May.
Article in English | MEDLINE | ID: mdl-38574829

ABSTRACT

Autophagy plays an important role in the lifecycle of viruses. However, there is currently a lack of systematic research on the relationship between Infectious Bronchitis Virus (IBV) and autophagy. This study aims to investigate the impact of IBV on autophagy and the role of autophagy in viral replication. We observed that IBV infection increased the expression of microtubule-associated protein 1 light chain 3, a marker of autophagy, decreased the expression of sequestosome 1, and led to elevated intracellular LC3 puncta levels. These findings suggest that IBV infection activates the autophagic process in cells. To investigate the impact of autophagy on the replication of IBV, we utilized rapamycin as an autophagy activator and 3-methyladenine as an autophagy inhibitor. Our results indicate that IBV promotes viral replication by inducing autophagy. Further investigation revealed that IBV induces autophagosome formation by inhibiting the mTOR-ULK1 pathway and activating the activity of vacuolar protein sorting 34 (VPS34), autophagy-related gene 14, and the Beclin-1 complex. VPS34 plays a crucial role in this process, as inhibiting VPS34 protein activity enhances cell proliferation after IBV infection. Additionally, inhibiting VPS34 significantly improves the survival rate of IBV-infected chicks, suppresses IBV replication in the kidney, and alleviates tracheal, lung, and kidney damage caused by IBV infection. In summary, IBV infection can induce autophagy by modulating the mTOR/ULK1 signaling pathway and activating the VPS34 complex, while autophagy serves to promote virus replication.


Subject(s)
Autophagy , Chickens , Class III Phosphatidylinositol 3-Kinases , Infectious bronchitis virus , Virus Replication , Infectious bronchitis virus/physiology , Animals , Class III Phosphatidylinositol 3-Kinases/metabolism , Chickens/virology , Coronavirus Infections/virology , Coronavirus Infections/metabolism , Sirolimus/pharmacology , Beclin-1/metabolism , Beclin-1/genetics , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Cell Line , Poultry Diseases/virology , Autophagosomes/metabolism , Autophagosomes/virology , Chlorocebus aethiops , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
2.
Cell ; 184(24): 5950-5969.e22, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34741801

ABSTRACT

The biogenesis of mammalian autophagosomes remains to be fully defined. Here, we used cellular and in vitro membrane fusion analyses to show that autophagosomes are formed from a hitherto unappreciated hybrid membrane compartment. The autophagic precursors emerge through fusion of FIP200 vesicles, derived from the cis-Golgi, with endosomally derived ATG16L1 membranes to generate a hybrid pre-autophagosomal structure, HyPAS. A previously unrecognized apparatus defined here controls HyPAS biogenesis and mammalian autophagosomal precursor membranes. HyPAS can be modulated by pharmacological agents whereas its formation is inhibited upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or by expression of SARS-CoV-2 nsp6. These findings reveal the origin of mammalian autophagosomal membranes, which emerge via convergence of secretory and endosomal pathways, and show that this process is targeted by microbial factors such as coronaviral membrane-modulating proteins.


Subject(s)
Autophagosomes/virology , COVID-19/virology , Autophagy , COVID-19/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endosomes/physiology , Endosomes/virology , Golgi Apparatus/physiology , HEK293 Cells , HeLa Cells , Humans , Membrane Fusion , Microscopy, Confocal , Phagosomes/metabolism , Phagosomes/virology , Qa-SNARE Proteins/biosynthesis , Receptors, sigma/biosynthesis , SARS-CoV-2 , Sarcoplasmic Reticulum Calcium-Transporting ATPases/biosynthesis , Synaptotagmins/biosynthesis , Sigma-1 Receptor
3.
Viruses ; 13(10)2021 10 09.
Article in English | MEDLINE | ID: mdl-34696464

ABSTRACT

Autophagic machinery is involved in selective and non-selective recruitment as well as degradation or exocytosis of cargoes, including pathogens. Dengue virus (DENV) infectioninduces autophagy that enhances virus replication and vesicle release to evade immune systemsurveillance. This study reveals that DENV2 induces autophagy in lung and liver cancer cells andshowed that DENV2 capsid, envelope, NS1, NS3, NS4B and host cell proinflammatory high mobilitygroup box 1 (HMGB1) proteins associated with autophagosomes which were purified by gradientcentrifugation. Capsid, NS1 and NS3 proteins showing high colocalization with LC3 protein in thecytoplasm of the infected cells were detected in the purified double-membrane autophagosome byimmunogold labeling under transmission electron microscopy. In DENV infected cells, the levels ofcapsid, envelope, NS1 and HMGB1 proteins are not significantly changed compared to the dramaticaccumulation of LC3-II and p62/SQSTM1 proteins when autophagic degradation was blocked bychloroquine, indicating that these proteins are not regulated by autophagic degradation machinery.We further demonstrated that purified autophagosomes were infectious when co-cultured withuninfected cells. Notably, these infectious autophagosomes contain DENV2 proteins, negativestrandand full-length genomic RNAs, but no viral particles. It is possible that the infectivity ofthe autophagosome originates from the full-length DENV RNA. Moreover, we reveal that DENV2promotes HMGB1 exocytosis partially through secretory autophagy. In conclusion, we are the firstto report that DENV2-induced double-membrane autophagosomes containing viral proteins andfull-length RNAs are infectious and not undergoing autophagic degradation. Our novel findingwarrants further validation of whether these intracellular vesicles undergo exocytosis to becomeinfectious autophagic vesicles.


Subject(s)
Autophagosomes/genetics , Autophagosomes/metabolism , Dengue Virus/genetics , A549 Cells , Animals , Autophagosomes/virology , Autophagy/genetics , Cell Line, Tumor , Chlorocebus aethiops , Dengue/virology , Genomics , HMGB1 Protein , Humans , Liver Neoplasms , RNA/metabolism , Vero Cells , Virion , Virus Replication
4.
Future Microbiol ; 16: 1135-1151, 2021 09.
Article in English | MEDLINE | ID: mdl-34468179

ABSTRACT

Coronaviruses quickly became a pandemic or epidemic, affecting large numbers of humans, due to their structural features and also because of their impacts on intracellular communications. The knowledge of the intracellular mechanism of virus distribution could help understand the coronavirus's proper effects on different pathways that lead to the infections. They protect themselves from recognition and damage the infected cell by using an enclosed membrane through hijacking the autophagy and endoplasmic reticulum-associated protein degradation pathways. The present study is a comprehensive review of the coronavirus strategy in upregulating the communication network of autophagy and endoplasmic reticulum-associated protein degradation.


Subject(s)
Autophagy , Coronavirus/pathogenicity , Endoplasmic Reticulum-Associated Degradation , Antiviral Agents/pharmacology , Autophagosomes/metabolism , Autophagosomes/virology , Autophagy/drug effects , COVID-19/metabolism , COVID-19/pathology , COVID-19/virology , Coronavirus/classification , Coronavirus Infections/drug therapy , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Coronavirus Infections/virology , Endoplasmic Reticulum-Associated Degradation/drug effects , Humans , SARS-CoV-2/pathogenicity , Virus Replication/drug effects , COVID-19 Drug Treatment
5.
Autophagy ; 17(7): 1768-1782, 2021 07.
Article in English | MEDLINE | ID: mdl-33890542

ABSTRACT

Despite the promising therapeutic effects of combinatory antiretroviral therapy (cART), 20% to 30% of HIV/AIDS patients living with long term infection still exhibit related cognitive and motor disorders. Clinical studies in HIV-infected patients revealed evidence of basal ganglia dysfunction, tremors, fine motor movement deficits, gait, balance, and increased risk of falls. Among older HIV+ adults, the frequency of cases with SNCA/α-synuclein staining is higher than in older healthy persons and may predict an increased risk of developing a neurodegenerative disease. The accumulation of SNCA aggregates known as Lewy Bodies is widely described to be directly linked to motor dysfunction. These aggregates are naturally removed by Macroautophagy/autophagy, a cellular housekeeping mechanism, that can be disturbed by HIV-1. The molecular mechanisms involved in linking HIV-1 proteins and autophagy remain mostly unclear and necessitates further exploration. We showed that HIV-1 Vpr protein triggers the accumulation of SNCA in neurons after decreasing lysosomal acidification, deregulating lysosome positioning, and the expression levels of several proteins involved in lysosomal maturation. Viruses and retroviruses such as HIV-1 are known to manipulate autophagy in order to use it for their replication while blocking the degradative final step, which could destroy the virus itself. Our study highlights how the suppression of neuronal autophagy by HIV-1 Vpr is a mechanism leading to toxic protein aggregation and neurodegeneration.Abbreviations: BLOC1: Biogenesis of Lysosome-related Organelles Complex 1; CART: combinatory antiretroviral therapy; CVB: coxsackievirus; DAPI: 4',6-diamidino-2-phenylindole; DENV: dengue virus; GFP: green fluorescent protein; HCV: hepatitis C virus; HCMV: human cytomegalovirus; HIV: human immunodeficiency virus; Env: HIV-1 envelope glycoproteins; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; VSV: Indiana vesiculovirus; LTR: Long Terminal Repeat; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MLBs: multilamellar bodies; RIPA: Radioimmunoprecipitation assay buffer; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; Tat: transactivator of TAR; TEM: transmission electron microscope; Vpr: Viral protein R.


Subject(s)
AIDS Dementia Complex/etiology , Lysosomes/virology , Neurons/virology , alpha-Synuclein/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , Animals , Autophagosomes/virology , Blotting, Western , Brain/pathology , Brain/virology , Fluorescent Antibody Technique , HIV-1 , Humans , Lysosomes/physiology , Macaca mulatta , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neurons/metabolism , Neurons/physiology
6.
Emerg Microbes Infect ; 10(1): 196-205, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33399028

ABSTRACT

ABSTRACT Following outbreaks of severe acute respiratory syndrome coronavirus (SARS-CoV) and the Middle East respiratory syndrome coronavirus (MERS-CoV) in 2002 and 2012, respectively, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the third highly pathogenic emerging human coronavirus (hCoV). SARS-CoV-2 is currently causing the global coronavirus disease 2019 (COVID-19) pandemic. CoV infections in target cells may stimulate the formation of numerous double-membrane autophagosomes and induce autophagy. Several studies provided evidence that hCoV infections are closely related to various cellular aspects associated with autophagy. Autophagy may even promote hCoV infection and replication. However, so far it is unclear how hCoV infections induce autophagy and whether the autophagic machinery is necessary for viral propagation. Here, we summarize the most recent advances concerning the mutual interplay between the autophagic machinery and the three emerging hCoVs, SARS-CoV, MERS-CoV, and SARS-CoV-2 and the model system mouse hepatitis virus. We also discuss the applicability of approved and well-tolerated drugs targeting autophagy as a potential treatment against COVID-19.


Subject(s)
Autophagosomes/virology , Autophagy , COVID-19/physiopathology , SARS-CoV-2/pathogenicity , Animals , Clinical Trials as Topic , Genome, Viral , Humans , Mice , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Murine hepatitis virus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , SARS-CoV-2/genetics , Virus Internalization/drug effects , COVID-19 Drug Treatment
7.
Autophagy ; 17(2): 476-495, 2021 02.
Article in English | MEDLINE | ID: mdl-32079455

ABSTRACT

Opportunistic bacterial infections amongst HIV-infected individuals contribute significantly to HIV-associated mortality. The role of HIV-mediated modulation of innate mechanisms like autophagy in promoting opportunistic infections, however, remains obscure. Here we show, HIV reactivation in or infection of macrophages inhibits autophagy and helps the survival of pathogenic Mycobacterium tuberculosis (Mtb) and nonpathogenic non-tuberculous mycobacterial strains (NTMs). The HIV-mediated impairment of xenophagy flux facilitated bacterial survival. Activation of autophagy by trehalose could induce xenophagy flux and kill intracellular Mtb or NTMs either during single or co-infections. Trehalose, we delineate, activates PIKFYVE leading to TFEB nuclear translocation in MCOLN1-dependent manner to induce autophagy. Remarkably, trehalose significantly reduced HIV-p24 levels in ex-vivo-infected PBMCs or PBMCs from treatment-naive HIV patients and also controlled mycobacterial survival within Mtb-infected animals. To conclude, we report leveraging of HIV-mediated perturbed host innate-immunity by opportunistic bacterial pathogens and show an attractive therapeutic strategy for HIV and associated co-morbidities.Abbreviations: AIDS: acquired immune deficiency syndrome; AMPK: AMP-activated protein kinase; ATG5: autophagy related 5; BafA1: bafilomycin A1; CFU: colony forming unit; CTSD: cathepsin D; CD63: CD63 molecule; EGFP: enhanced green fluorescent protein; FRET: Förster resonance energy transfer; GABARAP: gamma-aminobutyric acid receptor-associated protein; GAPDH: glyceraldehyde 3-phosphate dehydrogenase; GLUT: glucose transporter; HIV: human immunodeficiency virus; hMDMs: human monocyte derived macrophages; IL2: interleukin 2; LAMP1: lysosomal-associated membrane protein 1; LC3B-II: lipidated microtubule-associated proteins 1A/1B light chain 3B; Mtb: Mycobacterium tuberculosis; MTOR: mechanistic target of rapamycin; mRFP: monomeric red fluorescent protein; M6PR: mannose-6-phosphate receptor; NAC: N- acetyl- L -cysteine; NTM's: non-tuberculous mycobacteria; PBMC: Peripheral Blood Mononuclear cells; PIKFYVE: phosphoinositide kinase; FYVE-Type Zinc Finger; PHA: phytohemagglutinin; PMA: phorbol 12-myristate 13-acetate; PtdIns(3,5)P2: Phosphatidylinositol 3,5-bisphosphate; ptfLC3: pEGFP-mRFP-LC3; ROS: reactive oxygen species; SQSTM1: sequestosome1; TFEB: transcription factor EB; MCOLN1/TRPML1: mucolipin 1; PIP4P1/TMEM55B: Human trans-membrane Protein 55B; UVRAG: UV Radiation Resistance Associate; VPS35: vacuolar protein sorting associated protein 35; WDR45: WD repeat domain 45; YCAM: Yellow Chameleon.


Subject(s)
Autophagosomes/virology , Autophagy/drug effects , HIV Infections/drug therapy , Leukocytes, Mononuclear/drug effects , Trehalose/pharmacology , Animals , Autophagosomes/metabolism , Autophagy/physiology , Coinfection/drug therapy , Coinfection/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Macrophages/virology , Mycobacterium/metabolism , Mycobacterium/virology , Trehalose/metabolism
8.
Vet Microbiol ; 253: 108945, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33373883

ABSTRACT

Autophagy plays a momentous role in cellular responses against pathogens. However, the influence of the autophagy machinery on Muscovy duck reovirus (MDRV) infection is not yet confirmed. In this study, it was shown that MDRV infection significantly increased the number of autophagy-like vesicles in DF-1 cells under electron microscope and the LC3-I/LC3-II conversion, which was considered important indicators of autophagy. It was worth noting that the level of autophagy was positively correlated with MDRV replication. Further test results showed that MDRV-induced autophagy can promote virus replication in DF-1 cells, and both the envelope protein sigma A and non-structural protein sigma NS that play an important role in virus replication process can colocalize with the autophagosome marker molecule LC3-II by confocal immunofluorescence analysis. These results indicated that MDRV utilized the autophagosomes for replication. Through transfection of the dual fluorescent plasmid mcherry-EGFP-LC3 and fluorescence microscope observation, it was found that autophagosomes were more likely to fuse with lysosomes in MDRV-infected cells compared with the blank group. The phenomenon of pEGFP-LC3B fluorescent spot and LAMP1 co-localization appeared in MDRV infected cells, indicating that MDRV infection would promote the fusion of autophagosomes and the lysosomes. Conversely, accumulation of p62 was observed by immunoblotting, suggesting that autolysosomes does not exert effective degradation. MDRV infection triggered a incomplete autophagic response. Further studies found that the expression of LAMP1, a marker protein of late endosome/early lysosome, increased significantly in MDRV-infected cells, suggesting an increase in the number of immature lysosomes. In addition, the experiment detected the maturation of the lysosomal acid hydrolase Cathepsin D in the cells, and found that the expression of the 33 kDa mature form of Cathepsin D was significantly reduced after MDRV infection, indicating that MDRV inhibits the maturation of lysosomes. In general, MDRV infection induces autophagy of DF-1 cells, promotes the fusion of autophagosomes and lysosomes, inhibits autophagolysosome degradation, and promotes virus replication.


Subject(s)
Autophagosomes/virology , Autophagy , Lysosomes/metabolism , Orthoreovirus, Avian/physiology , Virus Replication , Animals , Cathepsin D/metabolism , Cell Line , Chickens , Ducks , Fibroblasts/virology , Lysosomes/virology , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/pathogenicity
9.
Nat Commun ; 11(1): 5189, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060596

ABSTRACT

Among the various host cellular processes that are hijacked by flaviviruses, few mechanisms have been described with regard to viral egress. Here we investigate how flaviviruses exploit Src family kinases (SFKs) for exit from infected cells. We identify Lyn as a critical component for secretion of Dengue and Zika infectious particles and their corresponding virus like particles (VLPs). Pharmacological inhibition or genetic depletion of the SFKs, Lyn in particular, block virus secretion. Lyn-/- cells are impaired in virus release and are rescued when reconstituted with wild-type Lyn, but not a kinase- or palmitoylation-deficient Lyn mutant. We establish that virus particles are secreted in two distinct populations - one as free virions and the other enclosed within membranes. Lyn is critical for the latter, which consists of proteolytically processed, infectious virus progenies within autophagosome-derived vesicles. This process depends on Ulk1, Rab GTPases and SNARE complexes implicated in secretory but not degradative autophagy and occur with significantly faster kinetics than the conventional secretory pathway. Our study reveals a previously undiscovered Lyn-dependent exit route of flaviviruses in LC3+ secretory organelles that enables them to evade circulating antibodies and might affect tissue tropism.


Subject(s)
Autophagosomes/metabolism , Autophagosomes/virology , Flavivirus/metabolism , src-Family Kinases/metabolism , Animals , Autophagy , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line , Chlorocebus aethiops , Dengue , Dengue Virus/metabolism , Host Microbial Interactions/physiology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , SNARE Proteins/metabolism , Secretory Pathway , Vero Cells , Virion/metabolism , Virus Release , Zika Virus/metabolism , Zika Virus Infection , rab GTP-Binding Proteins/metabolism , src-Family Kinases/genetics
10.
Sci Rep ; 10(1): 15234, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943650

ABSTRACT

Enterovirus A71 (EV-A71), which belongs to the family Picornaviridae, can invade the central nervous system (CNS) and cause severe CNS complications or death. The EV-A71 antigen has been detected in the neurons in the brains of humans who died from EV-A71 infection. However, the effect of EV-A71 infection on human neuronal cells remains poorly understood. Human neural stem cells (NSCs) and IMR-32 neuroblastoma cells were differentiated into neuronal cells for this study. Although the neuronal cells were permissive to EV-A71 infection, EV-A71 infection did not induce an obvious cytopathic effect on the neuronal cells. EV-A71 infection did not induce apoptosis in neuronal cells. However, autophagy and autophagic flux were induced in EV-A71-infected neuronal cells. The production of autophagosomes was shown to be important for EV-A71 viral RNA (vRNA) replication in neuronal cells.


Subject(s)
Enterovirus A, Human/physiology , Enterovirus A, Human/pathogenicity , Neurons/virology , Autophagosomes/virology , Autophagy/physiology , Caspases/metabolism , Cell Differentiation , Cells, Cultured , Cytopathogenic Effect, Viral/physiology , Enterovirus A, Human/genetics , Enzyme Activation , Host Microbial Interactions/physiology , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/virology , Neurons/metabolism , Neurons/pathology , RNA, Viral/biosynthesis , RNA, Viral/genetics , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL