Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(1): e0297262, 2024.
Article in English | MEDLINE | ID: mdl-38277395

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evades the innate immune machinery through multiple viral proteins, including nonstructural protein 1 (NSP1). While NSP1 is known to suppress translation of host mRNAs, the mechanisms underlying its immune evasion properties remain elusive. By integrating RNA-seq, ribosome footprinting, and ChIP-seq in A549 cells we found that NSP1 predominantly represses transcription of immune-related genes by favoring Histone 3 Lysine 9 dimethylation (H3K9me2). G9a/GLP H3K9 methyltransferase inhibitor UNC0638 restored expression of antiviral genes and restricted SARS-CoV-2 replication. Our multi-omics study unravels an epigenetic mechanism underlying host immune evasion by SARS-CoV-2 NSP1. Elucidating the factors involved in this phenomenon, may have implications for understanding and treating viral infections and other immunomodulatory diseases.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/genetics , COVID-19/immunology , COVID-19/virology , Epigenetic Repression , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Viral Nonstructural Proteins/metabolism
2.
iScience ; 26(3): 106169, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36785787

ABSTRACT

Beta-coronaviruses have emerged as a severe threat to global health. Undercovering the interplay between host and beta-coronaviruses is essential for understanding disease pathogenesis and developing efficient treatments. Here we report that the transcription factors TFEB and TFE3 translocate from the cytosol to the nucleus in response to beta-coronavirus infection by a mechanism that requires activation of calcineurin phosphatase. In the nucleus, TFEB and TFE3 bind to the promoter of multiple lysosomal and immune genes. Accordingly, MHV-induced upregulation of immune regulators is significantly decreased in TFEB/TFE3-depleted cells. Conversely, over-expression of either TFEB or TFE3 is sufficient to increase expression of several cytokines and chemokines. The reduced immune response observed in the absence of TFEB and TFE3 results in increased cellular survival of infected cells but also in reduced lysosomal exocytosis and decreased viral infectivity. These results suggest a central role of TFEB and TFE3 in cellular response to beta-coronavirus infection.

3.
Nat Commun ; 13(1): 5986, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36216808

ABSTRACT

Enteroviruses are non-enveloped positive-sense RNA viruses that cause diverse diseases in humans. Their rapid multiplication depends on remodeling of cytoplasmic membranes for viral genome replication. It is unknown how virions assemble around these newly synthesized genomes and how they are then loaded into autophagic membranes for release through secretory autophagy. Here, we use cryo-electron tomography of infected cells to show that poliovirus assembles directly on replication membranes. Pharmacological untethering of capsids from membranes abrogates RNA encapsidation. Our data directly visualize a membrane-bound half-capsid as a prominent virion assembly intermediate. Assembly progression past this intermediate depends on the class III phosphatidylinositol 3-kinase VPS34, a key host-cell autophagy factor. On the other hand, the canonical autophagy initiator ULK1 is shown to restrict virion production since its inhibition leads to increased accumulation of virions in vast intracellular arrays, followed by an increased vesicular release at later time points. Finally, we identify multiple layers of selectivity in virus-induced autophagy, with a strong selection for RNA-loaded virions over empty capsids and the segregation of virions from other types of autophagosome contents. These findings provide an integrated structural framework for multiple stages of the poliovirus life cycle.


Subject(s)
Enterovirus Infections , Poliovirus , Autophagy , Capsid , Class III Phosphatidylinositol 3-Kinases , Humans , Poliovirus/genetics , RNA , Virion/genetics , Virus Assembly/physiology
4.
Environ Sci Technol ; 56(12): 8475-8484, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35653550

ABSTRACT

Recent discovery of vesicle-cloaked virus clusters (i.e., viral vesicles) has greatly challenged the central paradigm of viral transmission and infection as a single virion. To understand the environmental transmission of viral vesicles, we used an in vivo model to investigate their environmental persistence and engineering control by disinfection. Murine rotavirus vesicles maintained both their integrity and infectivity after incubation in filtered freshwater and wastewater for at least 7 days, with 24.5-27.5% of the vesicles still intact at 16 weeks after exposure to both waters. Free chlorine disinfection at a dosage of 13.3 mg min L-1 did not decompose murine rotavirus vesicles, and it was much less effective in inactivating rotaviruses inside vesicles than free rotaviruses based on the quantification of rotavirus shedding in mouse stool and rotavirus replication in small intestines. Rotavirus vesicles may be more environmentally transmissible than free rotaviruses regardless of disinfection. Vesicle-mediated en bloc transmission could be responsible for vesicles' resistance to disinfection due to an increased multiplicity of infection and/or genetic recombination or reassortment to promote infection. Our work highlights the environmental, biological, and public health significance of viral vesicles, and the findings call for urgent action in advancing disinfection for pathogen control.


Subject(s)
Rotavirus , Animals , Chlorine/pharmacology , Disinfection , Feces , Mice , Rotavirus/genetics , Wastewater
5.
Environ Sci Technol ; 56(9): 5381-5389, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35434991

ABSTRACT

Waterborne human pathogenic viruses challenge global health and economy. Viruses were long believed to transmit among hosts as individual, free particles. However, recent evidence indicates that viruses also transmit in populations, so-called en bloc transmission, by either interacting with coexisting bacteria, free-living amoebas, and other higher organisms through endosymbiosis and surface binding, or by being clustered inside membrane-bound vesicles or simply self-aggregating with themselves. En bloc transmission of viruses and virus-microbiome interactions could enable viruses to enhance their infectivity, increase environmental persistence, and resist inactivation from disinfection. Overlooking this type of transmission and virus-microbiome interactions may underestimate the environmental and public health risks of the viruses. We herein provide a critical perspective on waterborne human pathogenic viruses in complex microbial communities to elucidate the environmental implication of virus-microbiome interactions on virus infectivity, persistence, and disinfection. This perspective also provides insights on advancing disinfection and sanitation guidelines and regulations to protect the public health.


Subject(s)
Microbiota , Viruses , Bacteria , Disinfection , Humans
6.
Annu Rev Cell Dev Biol ; 37: 171-197, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34270326

ABSTRACT

Viral egress and transmission have long been described to take place through single free virus particles. However, viruses can also shed into the environment and transmit as populations clustered inside extracellular vesicles (EVs), a process we had first called vesicle-mediated en bloc transmission. These membrane-cloaked virus clusters can originate from a variety of cellular organelles including autophagosomes, plasma membrane, and multivesicular bodies. Their viral cargo can be multiples of nonenveloped or enveloped virus particles or even naked infectious genomes, but egress is always nonlytic, with the cell remaining intact. Here we put forth the thesis that EV-cloaked viral clusters are a distinct form of infectious unit as compared to free single viruses (nonenveloped or enveloped) or even free virus aggregates. We discuss how efficient and prevalent these infectious EVs are in the context of virus-associated diseases and highlight the importance of their proper detection and disinfection for public health.


Subject(s)
Extracellular Vesicles , Viruses , Extracellular Vesicles/metabolism , Viruses/genetics
7.
Mol Cell ; 81(10): 2061-2063, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34019787

ABSTRACT

Jones et al. (2021) and Zhang et al. (2021) reveal by cryo-EM the oligomeric crown-like structure formed by a membrane-associated Chikungunya virus replication protein that gates the export of newly synthesized viral RNA from viral replication organelles.


Subject(s)
Chikungunya virus , Virus Replication , Chikungunya virus/genetics , Organelles , RNA, Viral/genetics
8.
J Biol Chem ; 297(1): 100813, 2021 07.
Article in English | MEDLINE | ID: mdl-34023384

ABSTRACT

Niemann-Pick C (NPC) is an autosomal recessive disorder characterized by mutations in the NPC1 or NPC2 genes encoding endolysosomal lipid transport proteins, leading to cholesterol accumulation and autophagy dysfunction. We have previously shown that enrichment of NPC1-deficient cells with the anionic lipid lysobisphosphatidic acid (LBPA; also called bis(monoacylglycerol)phosphate) via treatment with its precursor phosphatidylglycerol (PG) results in a dramatic decrease in cholesterol storage. However, the mechanisms underlying this reduction are unknown. In the present study, we showed using biochemical and imaging approaches in both NPC1-deficient cellular models and an NPC1 mouse model that PG incubation/LBPA enrichment significantly improved the compromised autophagic flux associated with NPC1 disease, providing a route for NPC1-independent endolysosomal cholesterol mobilization. PG/LBPA enrichment specifically enhanced the late stages of autophagy, and effects were mediated by activation of the lysosomal enzyme acid sphingomyelinase. PG incubation also led to robust and specific increases in LBPA species with polyunsaturated acyl chains, potentially increasing the propensity for membrane fusion events, which are critical for late-stage autophagy progression. Finally, we demonstrated that PG/LBPA treatment efficiently cleared cholesterol and toxic protein aggregates in Purkinje neurons of the NPC1I1061T mouse model. Collectively, these findings provide a mechanistic basis supporting cellular LBPA as a potential new target for therapeutic intervention in NPC disease.


Subject(s)
Autophagy , Cholesterol/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Lysophospholipids/metabolism , Lysosomes/metabolism , Monoglycerides/metabolism , Animals , Autophagy/drug effects , Endosomes/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells , Homeostasis/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mutation/genetics , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Phosphatidylglycerols/pharmacology , Purkinje Cells/drug effects , Purkinje Cells/metabolism , Sequestosome-1 Protein/metabolism , Sphingomyelin Phosphodiesterase/metabolism
9.
Environ Sci Technol ; 55(9): 6197-6205, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33856208

ABSTRACT

An individual virion was long believed to act as an independent infectious unit in virology, until the recent discovery of vesicle-cloaked virus clusters which has greatly challenged this central paradigm. Vesicle-cloaked virus clusters (also known as viral vesicles) are phospholipid-bilayer encapsulated fluid sacs that contain multiple virions or multiple copies of viral genomes. Norovirus is a global leading causative agent of gastroenteritis, and the reported prevalence of vesicle-cloaked norovirus clusters in stool has raised concerns whether the current disinfection, sanitation, and hygiene practices can effectively control environmental pollution by these pathogenic units. In this study, we have demonstrated that vesicle-cloaked murine norovirus (MNV-1) clusters were highly persistent under temperature variation (i.e., freeze-thaw) and they were partially resistant to detergent decomposition. MNV-1 vesicles were 1.89-3.17-fold more infectious in vitro than their free virus counterparts. Most importantly, MNV-1 vesicles were up to 2.16-times more resistant to UV254 disinfection than free MNV-1 at a low viral load in vitro. Interestingly, with the increase of the viral load, free MNV-1 and MNV-1 vesicles showed equivalent resistance to UV254 disinfection. We show that the increased multiplicity of infection provided by vesicles is in part responsible for these attributes. Our study, for the first time, sheds light on the environmental behavior of vesicle-cloaked virus clusters as unique emerging pathogenic units. Our study highlights the need to revisit current paradigms of disinfection, sanitation, and hygiene practices for protecting public health.


Subject(s)
Caliciviridae Infections , Norovirus , Animals , Disinfection , Feces , Mice
11.
Mucosal Immunol ; 14(4): 937-948, 2021 07.
Article in English | MEDLINE | ID: mdl-33731830

ABSTRACT

Microbial translocation contributes to persistent inflammation in both treated and untreated HIV infection. Although translocation is due in part to a disintegration of the intestinal epithelial barrier, there is a bias towards the translocation of Proteobacteria. We hypothesized that intestinal epithelial microvesicle cargo differs after HIV infection and contributes to biased translocation. We isolated gastrointestinal luminal microvesicles before and after progressive simian immunodeficiency virus (SIV) infection in rhesus macaques and measured miRNA and antimicrobial peptide content. We demonstrate that these microvesicles display decreased miR-28-5p, -484, -584-3p, and -584-5p, and let-7b-3p, as well as increased beta-defensin 1 after SIV infection. We further observed dose-dependent growth sensitivity of commensal Lactobacillus salivarius upon co-culture with isolated microvesicles. Infection-associated microvesicle differences were not mirrored in non-progressively SIV-infected sooty mangabeys. Our findings describe novel alterations of antimicrobial control after progressive SIV infection that influence the growth of translocating bacterial taxa. These studies may lead to the development of novel therapeutics for treating chronic HIV infection, microbial translocation, and inflammation.


Subject(s)
Bacterial Translocation , Dysbiosis/etiology , Extracellular Vesicles/metabolism , Gastrointestinal Microbiome , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus , Animals , Biomarkers , Disease Progression , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Macaca mulatta , MicroRNAs/genetics , Simian Acquired Immunodeficiency Syndrome/complications
12.
Autophagy ; 17(1): 1-382, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33634751

ABSTRACT

In 2008, we published the first set of guidelines for standardizing research in autophagy. Since then, this topic has received increasing attention, and many scientists have entered the field. Our knowledge base and relevant new technologies have also been expanding. Thus, it is important to formulate on a regular basis updated guidelines for monitoring autophagy in different organisms. Despite numerous reviews, there continues to be confusion regarding acceptable methods to evaluate autophagy, especially in multicellular eukaryotes. Here, we present a set of guidelines for investigators to select and interpret methods to examine autophagy and related processes, and for reviewers to provide realistic and reasonable critiques of reports that are focused on these processes. These guidelines are not meant to be a dogmatic set of rules, because the appropriateness of any assay largely depends on the question being asked and the system being used. Moreover, no individual assay is perfect for every situation, calling for the use of multiple techniques to properly monitor autophagy in each experimental setting. Finally, several core components of the autophagy machinery have been implicated in distinct autophagic processes (canonical and noncanonical autophagy), implying that genetic approaches to block autophagy should rely on targeting two or more autophagy-related genes that ideally participate in distinct steps of the pathway. Along similar lines, because multiple proteins involved in autophagy also regulate other cellular pathways including apoptosis, not all of them can be used as a specific marker for bona fide autophagic responses. Here, we critically discuss current methods of assessing autophagy and the information they can, or cannot, provide. Our ultimate goal is to encourage intellectual and technical innovation in the field.


Subject(s)
Autophagy , Animals , Autophagosomes , Autophagy/physiology , Autophagy-Related Proteins/metabolism , Biological Assay/standards , Biomarkers , Humans , Lysosomes
13.
Cell ; 183(6): 1520-1535.e14, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33157038

ABSTRACT

ß-Coronaviruses are a family of positive-strand enveloped RNA viruses that includes the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Much is known regarding their cellular entry and replication pathways, but their mode of egress remains uncertain. Using imaging methodologies and virus-specific reporters, we demonstrate that ß-coronaviruses utilize lysosomal trafficking for egress rather than the biosynthetic secretory pathway more commonly used by other enveloped viruses. This unconventional egress is regulated by the Arf-like small GTPase Arl8b and can be blocked by the Rab7 GTPase competitive inhibitor CID1067700. Such non-lytic release of ß-coronaviruses results in lysosome deacidification, inactivation of lysosomal degradation enzymes, and disruption of antigen presentation pathways. ß-Coronavirus-induced exploitation of lysosomal organelles for egress provides insights into the cellular and immunological abnormalities observed in patients and suggests new therapeutic modalities.


Subject(s)
COVID-19/metabolism , SARS-CoV-2/metabolism , Secretory Pathway , Virus Release , ADP-Ribosylation Factors/metabolism , Animals , COVID-19/pathology , Female , HeLa Cells , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Lysosomes , Mice , Thiourea/analogs & derivatives , Thiourea/pharmacology , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins , COVID-19 Drug Treatment
14.
Nat Commun ; 11(1): 2968, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32528015

ABSTRACT

Human noroviruses are the leading cause of severe childhood diarrhea worldwide, yet we know little about their pathogenic mechanisms. Murine noroviruses cause diarrhea in interferon-deficient adult mice but these hosts also develop systemic pathology and lethality, reducing confidence in the translatability of findings to human norovirus disease. Herein we report that a murine norovirus causes self-resolving diarrhea in the absence of systemic disease in wild-type neonatal mice, thus mirroring the key features of human norovirus disease and representing a norovirus small animal disease model in wild-type mice. Intriguingly, lymphocytes are critical for controlling acute norovirus replication while simultaneously contributing to disease severity, likely reflecting their dual role as targets of viral infection and key components of the host response.


Subject(s)
Caliciviridae Infections/pathology , Diarrhea/virology , Norovirus/pathogenicity , Animals , Animals, Newborn , Disease Models, Animal , Female , Lymphocytes/metabolism , Male , Mice , Mice, Inbred BALB C
15.
Cell Rep ; 28(4): 923-937.e3, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31340154

ABSTRACT

In cancer biology, the functional interpretation of genomic alterations is critical to achieve the promise of genomic profiling in the clinic. For chronic lymphocytic leukemia (CLL), a heterogeneous disease of B-lymphocytes maturing under constitutive B cell receptor (BCR) stimulation, the functional role of diverse clonal mutations remains largely unknown. Here, we demonstrate that alterations in BCR signaling dynamics underlie the progression of B cells toward malignancy. We reveal emergent dynamic features-bimodality, hypersensitivity, and hysteresis-in the BCR signaling pathway of primary CLL B cells. These signaling abnormalities in CLL quantitatively derive from BCR clustering and constitutive signaling with positive feedback reinforcement, as demonstrated through single-cell analysis of phospho-responses, computational modeling, and super-resolution imaging. Such dysregulated signaling segregates CLL patients by disease severity and clinical presentation. These findings provide a quantitative framework and methodology to assess complex and heterogeneous leukemia pathology and to inform therapeutic strategies in parallel with genomic profiling.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Adult , Aged , Aged, 80 and over , Biophysical Phenomena , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Feedback, Physiological/drug effects , Female , Humans , Male , Middle Aged , Models, Biological , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , Signal Transduction/drug effects , Single-Cell Analysis , Small Molecule Libraries/pharmacology
16.
mBio ; 10(3)2019 05 28.
Article in English | MEDLINE | ID: mdl-31138755

ABSTRACT

Extracellular vesicles (EVs) are major vehicles for transporting viruses en bloc among hosts. While RNA viruses make up the great majority of transmission by EVs, in a recent article in mBio (mBio 10:e00379-19, 2019, https://mbio.asm.org/content/10/2/e00379-19.long), Morris-Love and colleagues revealed that a double-stranded DNA (dsDNA) virus, JC polyomavirus (JCPyV), a major cause of progressive multifocal leukoencephalopathy (PML), can be released from and transmitted to other glia in EVs. This mode of transmission appears to be highly infectious, independent of the free virus attachment and entry receptors LSTc and 5-HT2, and protected from neutralizing antibodies. This novel form of JCPyV transmission may potentially explain its dissemination into the central nervous system (CNS) and its increased virulence.


Subject(s)
Extracellular Vesicles , JC Virus , Leukoencephalopathy, Progressive Multifocal , Humans , Neuroglia , Virus Attachment
17.
Virus Res ; 265: 143-149, 2019 05.
Article in English | MEDLINE | ID: mdl-30928427

ABSTRACT

En Bloc transmission of viruses allow multiple genomes to collectivelly penetrate and initiate infection in the same cell, often resulting in enhanced infectivity. Given the quasispecies (mutant cloud) nature of RNA viruses and many DNA viruses, en bloc transmission of multiple genomes provides different starting points in sequence space to initiate adaptive walks, and has implications for modulation of viral fitness and for the response of viral populations to lethal mutagenesis. Mechanisms that can enable multiple viral genomes to be transported en bloc among hosts has only recently been gaining attention. A growing body of research suggests that extracellular vesicles (EV) are highly prevalent and robust vehicles for en bloc delivery of viral particles and naked infectious genomes among organisms. Both RNA and DNA viruses appear to exploit these vesicles to increase their multiplicity of infection and enhance virulence.


Subject(s)
Extracellular Vesicles/virology , Genome, Viral , Virus Diseases/transmission , Viruses/genetics , Animals , DNA Viruses/genetics , DNA Viruses/pathogenicity , Humans , Membrane Lipids/metabolism , Mice , Quasispecies , RNA Viruses/genetics , RNA Viruses/pathogenicity , Virion/genetics , Virus Replication , Viruses/pathogenicity
18.
Cell Host Microbe ; 24(2): 208-220.e8, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30092198

ABSTRACT

In enteric viral infections, such as those with rotavirus and norovirus, individual viral particles shed in stool are considered the optimal units of fecal-oral transmission. We reveal that rotaviruses and noroviruses are also shed in stool as viral clusters enclosed within vesicles that deliver a high inoculum to the receiving host. Cultured cells non-lytically release rotaviruses and noroviruses inside extracellular vesicles. In addition, stools of infected hosts contain norovirus and rotavirus within vesicles of exosomal or plasma membrane origin. These vesicles remain intact during fecal-oral transmission and thereby transport multiple viral particles collectively to the next host, enhancing both the MOI and disease severity. Vesicle-cloaked viruses are non-negligible populations in stool and have a disproportionately larger contribution to infectivity than free viruses. Our findings indicate that vesicle-cloaked viruses are highly virulent units of fecal-oral transmission and highlight a need for antivirals targeting vesicles and virus clustering.


Subject(s)
Caliciviridae Infections/transmission , Extracellular Vesicles/virology , Feces/virology , Rotavirus Infections/transmission , Animals , Caliciviridae Infections/virology , Child, Preschool , Disease Transmission, Infectious , Exosomes/virology , Female , Humans , Male , Mice, Inbred BALB C , Norovirus/genetics , Norovirus/pathogenicity , Rotavirus/genetics , Rotavirus/pathogenicity , Rotavirus Infections/virology , Swine , Virus Shedding
19.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29950420

ABSTRACT

Following the initial detection of viral infection, innate immune responses trigger the induction of numerous interferon-stimulated genes (ISGs) to inhibit virus replication and dissemination. One such ISG encodes cholesterol-25-hydroxylase (CH25H), an enzyme that catalyzes the oxidation of cholesterol to form a soluble product, 25-hydroxycholesterol (25HC). Recent studies have found that CH25H is broadly antiviral; it inhibits infection by several viruses. For enveloped viruses, 25HC inhibits membrane fusion, likely by altering membrane characteristics such as hydrophobicity or cholesterol aggregation. However, the mechanisms by which 25HC restricts infection of nonenveloped viruses are unknown. We examined whether 25HC restricts infection by mammalian reovirus. Treatment with 25HC restricted infection by reovirus prototype strains type 1 Lang and type 3 Dearing. In contrast to reovirus virions, 25HC did not restrict infection by reovirus infectious subvirion particles (ISVPs), which can penetrate either directly at the cell surface or in early endosomal membranes. Treatment with 25HC altered trafficking of reovirus particles to late endosomes and delayed the kinetics of reovirus uncoating. These results suggest that 25HC inhibits the efficiency of cellular entry of reovirus virions, which may require specific endosomal membrane dynamics for efficient membrane penetration.IMPORTANCE The innate immune system is crucial for effective responses to viral infection. Type I interferons, central components of innate immunity, induce expression of hundreds of ISGs; however, the mechanisms of action of these antiviral proteins are not well understood. CH25H, encoded by an ISG, represents a significant constituent of these cellular antiviral strategies, as its metabolic product, 25HC, can act in both an autocrine and a paracrine fashion to protect cells from infection and has been shown to limit viral infection in animal models. Further investigation into the mechanism of action of 25HC may inform novel antiviral therapies and influence the use of mammalian reovirus in clinical trials as an oncolytic agent.


Subject(s)
Antiviral Agents/pharmacology , Hydroxycholesterols/metabolism , Hydroxycholesterols/pharmacology , Reoviridae/drug effects , Steroid Hydroxylases/genetics , Virus Replication/drug effects , Animals , Cell Line , HeLa Cells , Humans , Immunity, Innate/drug effects , Interferon Type I/genetics , Interferon Type I/pharmacology , Reoviridae/physiology , Steroid Hydroxylases/metabolism , Virion , Virus Internalization/drug effects
20.
PLoS Pathog ; 14(3): e1006916, 2018 03.
Article in English | MEDLINE | ID: mdl-29538454

ABSTRACT

Entry of hepatitis C virus (HCV) into hepatocytes is a complex process that involves numerous cellular factors, including the scavenger receptor class B type 1 (SR-B1), the tetraspanin CD81, and the tight junction (TJ) proteins claudin-1 (CLDN1) and occludin (OCLN). Despite expression of all known HCV-entry factors, in vitro models based on hepatoma cell lines do not fully reproduce the in vivo susceptibility of liver cells to primary HCV isolates, implying the existence of additional host factors which are critical for HCV entry and/or replication. Likewise, HCV replication is severely impaired within hepatocellular carcinoma (HCC) tissue in vivo, but the mechanisms responsible for this restriction are presently unknown. Here, we identify tumor-associated calcium signal transducer 2 (TACSTD2), one of the most downregulated genes in primary HCC tissue, as a host factor that interacts with CLDN1 and OCLN and regulates their cellular localization. TACSTD2 gene silencing disrupts the typical linear distribution of CLDN1 and OCLN along the cellular membrane in both hepatoma cells and primary human hepatocytes, recapitulating the pattern observed in vivo in primary HCC tissue. Mechanistic studies suggest that TACSTD2 is involved in the phosphorylation of CLDN1 and OCLN, which is required for their proper cellular localization. Silencing of TACSTD2 dramatically inhibits HCV infection with a pan-genotype effect that occurs at the level of viral entry. Our study identifies TACSTD2 as a novel regulator of two major HCV-entry factors, CLDN1 and OCLN, which is strongly downregulated in malignant hepatocytes. These results provide new insights into the complex process of HCV entry into hepatocytes and may assist in the development of more efficient cellular systems for HCV propagation in vitro.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Hepatocellular/virology , Cell Adhesion Molecules/metabolism , Claudin-1/metabolism , Hepacivirus/pathogenicity , Hepatitis C/virology , Liver Neoplasms/virology , Occludin/metabolism , Antigens, Neoplasm/genetics , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/genetics , Claudin-1/genetics , Down-Regulation , Hepatitis C/complications , Hepatitis C/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Humans , Liver Neoplasms/epidemiology , Liver Neoplasms/metabolism , Occludin/genetics , Virus Internalization , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL