Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
PLoS Pathog ; 20(9): e1012509, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39241103

ABSTRACT

The replication organelle of hepatitis C virus (HCV), called membranous web, is derived from the endoplasmic reticulum (ER) and mainly comprises double membrane vesicles (DMVs) that concentrate the viral replication complexes. It also tightly associates with lipid droplets (LDs), which are essential for virion morphogenesis. In particular acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1), a rate-limiting enzyme in triglyceride synthesis, promotes early steps of virus assembly. The close proximity between ER membranes, DMVs and LDs therefore permits the efficient coordination of the HCV replication cycle. Here, we demonstrate that exaggerated LD accumulation due to the excessive expression of the DGAT1 isozyme, DGAT2, dramatically impairs the formation of the HCV membranous web. This effect depended on the enzymatic activity and ER association of DGAT2, whereas the mere LD accumulation was not sufficient to hamper HCV RNA replication. Our lipidomics data indicate that both HCV infection and DGAT2 overexpression induced membrane lipid biogenesis and markedly increased phospholipids with long chain polyunsaturated fatty acids, suggesting a dual use of these lipids and their possible competition for LD and DMV biogenesis. On the other hand, overexpression of DGAT2 depleted specific phospholipids, particularly oleyl fatty acyl chain-containing phosphatidylcholines, which, in contrast, are increased in HCV-infected cells and likely essential for viral infection. In conclusion, our results indicate that lipid exchanges occurring during LD biogenesis regulate the composition of intracellular membranes and thereby affect the formation of the HCV replication organelle. The potent antiviral effect observed in our DGAT2 overexpression system unveils lipid flux that may be relevant in the context of steatohepatitis, a hallmark of HCV infection, but also in physiological conditions, locally in specific subdomains of the ER membrane. Thus, LD formation mediated by DGAT1 and DGAT2 might participate in the spatial compartmentalization of HCV replication and assembly factories within the membranous web.


Subject(s)
Diacylglycerol O-Acyltransferase , Endoplasmic Reticulum , Hepacivirus , Triglycerides , Virus Replication , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/genetics , Humans , Hepacivirus/physiology , Virus Replication/physiology , Triglycerides/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Hepatitis C/metabolism , Hepatitis C/virology , Lipid Droplets/metabolism , Lipid Droplets/virology
2.
J Biol Chem ; 300(5): 107286, 2024 May.
Article in English | MEDLINE | ID: mdl-38636657

ABSTRACT

Hepatitis C virus (HCV) infection is tightly connected to the lipid metabolism with lipid droplets (LDs) serving as assembly sites for progeny virions. A previous LD proteome analysis identified annexin A3 (ANXA3) as an important HCV host factor that is enriched at LDs in infected cells and required for HCV morphogenesis. To further characterize ANXA3 function in HCV, we performed proximity labeling using ANXA3-BioID2 as bait in HCV-infected cells. Two of the top proteins identified proximal to ANXA3 during HCV infection were the La-related protein 1 (LARP1) and the ADP ribosylation factor-like protein 8B (ARL8B), both of which have been previously described to act in HCV particle production. In follow-up experiments, ARL8B functioned as a pro-viral HCV host factor without localizing to LDs and thus likely independent of ANXA3. In contrast, LARP1 interacts with HCV core protein in an RNA-dependent manner and is translocated to LDs by core protein. Knockdown of LARP1 decreased HCV spreading without altering HCV RNA replication or viral titers. Unexpectedly, entry of HCV particles and E1/E2-pseudotyped lentiviral particles was reduced by LARP1 depletion, whereas particle production was not altered. Using a recombinant vesicular stomatitis virus (VSV)ΔG entry assay, we showed that LARP1 depletion also decreased entry of VSV with VSV, MERS, and CHIKV glycoproteins. Therefore, our data expand the role of LARP1 as an HCV host factor that is most prominently involved in the early steps of infection, likely contributing to endocytosis of viral particles through the pleiotropic effect LARP1 has on the cellular translatome.


Subject(s)
Annexin A3 , Hepacivirus , Hepatitis C , SS-B Antigen , Virus Internalization , Humans , Annexin A3/metabolism , Annexin A3/genetics , Autoantigens/metabolism , Autoantigens/genetics , HEK293 Cells , Hepacivirus/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatitis C/genetics , Host-Pathogen Interactions , Lipid Droplets/metabolism , Lipid Droplets/virology , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Viral Core Proteins/metabolism , Viral Core Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics
3.
Plant J ; 119(5): 2484-2499, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39007841

ABSTRACT

Pathogen infection induces massive reprogramming of host primary metabolism. Lipid and fatty acid (FA) metabolism is generally disrupted by pathogens and co-opted for their proliferation. Lipid droplets (LDs) that play important roles in regulating cellular lipid metabolism are utilized by a variety of pathogens in mammalian cells. However, the function of LDs during pathogenic infection in plants remains unknown. We show here that infection by rice black streaked dwarf virus (RBSDV) affects the lipid metabolism of maize, which causes elevated accumulation of C18 polyunsaturated fatty acids (PUFAs) leading to viral proliferation and symptom development. The overexpression of one of the two novel LD-associated proteins (LDAPs) of maize (ZmLDAP1 and ZmLDAP2) induces LD clustering. The core capsid protein P8 of RBSDV interacts with ZmLDAP2 and prevents its degradation through the ubiquitin-proteasome system mediated by a UBX domain-containing protein, PUX10. In addition, silencing of ZmLDAP2 downregulates the expression of FA desaturase genes in maize, leading to a decrease in C18 PUFAs levels and suppression of RBSDV accumulation. Our findings reveal that plant virus may recruit LDAP to regulate cellular FA metabolism to promote viral multiplication and infection. These results expand the knowledge of LD functions and viral infection mechanisms in plants.


Subject(s)
Fatty Acids , Plant Diseases , Plant Proteins , Virus Replication , Zea mays , Zea mays/virology , Zea mays/metabolism , Zea mays/genetics , Plant Proteins/metabolism , Plant Proteins/genetics , Plant Diseases/virology , Fatty Acids/metabolism , Lipid Metabolism , Lipid Droplet Associated Proteins/metabolism , Lipid Droplet Associated Proteins/genetics , Lipid Droplets/metabolism , Lipid Droplets/virology , Plant Viruses/physiology , Gene Expression Regulation, Plant , Reoviridae/physiology
4.
PLoS Pathog ; 17(4): e1009496, 2021 04.
Article in English | MEDLINE | ID: mdl-33872335

ABSTRACT

LINE-1 (L1) retrotransposons are autonomous transposable elements that can affect gene expression and genome integrity. Potential consequences of exogenous viral infections for L1 activity have not been studied to date. Here, we report that hepatitis C virus (HCV) infection causes a significant increase of endogenous L1-encoded ORF1 protein (L1ORF1p) levels and translocation of L1ORF1p to HCV assembly sites at lipid droplets. HCV replication interferes with retrotransposition of engineered L1 reporter elements, which correlates with HCV RNA-induced formation of stress granules and can be partially rescued by knockdown of the stress granule protein G3BP1. Upon HCV infection, L1ORF1p localizes to stress granules, associates with HCV core in an RNA-dependent manner and translocates to lipid droplets. While HCV infection has a negative effect on L1 mobilization, L1ORF1p neither restricts nor promotes HCV infection. In summary, our data demonstrate that HCV infection causes an increase of endogenous L1 protein levels and that the observed restriction of retrotransposition of engineered L1 reporter elements is caused by sequestration of L1ORF1p in HCV-induced stress granules.


Subject(s)
Carcinoma, Hepatocellular/virology , DNA Helicases/metabolism , Hepacivirus/physiology , Hepatitis C/virology , Liver Neoplasms/virology , Long Interspersed Nucleotide Elements/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Ribonucleoproteins/metabolism , Cell Line, Tumor , Cytoplasmic Granules/virology , DNA Helicases/genetics , Humans , Lipid Droplets/virology , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , Ribonucleoproteins/genetics
5.
Mem Inst Oswaldo Cruz ; 118: e230044, 2023.
Article in English | MEDLINE | ID: mdl-37820117

ABSTRACT

Lipid droplets (LD) are evolutionarily conserved lipid-enriched organelles with a diverse array of cell- and stimulus-regulated proteins. Accumulating evidence demonstrates that intracellular pathogens exploit LD as energy sources, replication sites, and part of the mechanisms of immune evasion. Nevertheless, LD can also favor the host as part of the immune and inflammatory response to pathogens. The functions of LD in the central nervous system have gained great interest due to their presence in various cell types in the brain and for their suggested involvement in neurodevelopment and neurodegenerative diseases. Only recently have the roles of LD in neuroinfections begun to be explored. Recent findings reveal that lipid remodelling and increased LD biogenesis play important roles for Zika virus (ZIKV) replication and pathogenesis in neural cells. Moreover, blocking LD formation by targeting DGAT-1 in vivo inhibited virus replication and inflammation in the brain. Therefore, targeting lipid metabolism and LD biogenesis may represent potential strategies for anti-ZIKV treatment development. Here, we review the progress in understanding LD functions in the central nervous system in the context of the host response to Zika infection.


Subject(s)
Central Nervous System Infections , Lipid Droplets , Zika Virus Infection , Zika Virus , Humans , Lipid Droplets/metabolism , Lipid Droplets/physiology , Lipid Droplets/virology , Lipids/physiology , Virus Replication/physiology , Zika Virus/physiology , Zika Virus Infection/physiopathology , Zika Virus Infection/virology , Central Nervous System Infections/physiopathology , Central Nervous System Infections/virology
6.
Traffic ; 21(8): 545-551, 2020 08.
Article in English | MEDLINE | ID: mdl-32424954

ABSTRACT

Positive sense (+) RNA viruses exploit membranes from a variety of cellular organelles to support the amplification of their genomes. This association concurs with the formation of vesicles whose main morphological feature is that of being wrapped by a double membrane. In the case of the SARS-CoV virus, the outer membrane is not discrete for each vesicle, but seems to be continuous and shared between many individual vesicles, a difference with other +RNA viruses whose nature has remained elusive. I present morphological, biochemical and pharmacological arguments defending the striking analogy of this arrangement and that of entangled, nascent Lipid Droplets whose birth has been aborted by an excess of Phosphatidic Acid. Since Phosphatidic Acid can be targeted with therapeutical purposes, considering this working hypothesis may prove important in tackling SARS-CoV infection.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Models, Biological , Phosphatidic Acids/metabolism , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Betacoronavirus/pathogenicity , COVID-19 , Host Microbial Interactions/physiology , Humans , Lipid Droplets/metabolism , Lipid Droplets/virology , Pandemics , SARS-CoV-2 , Virus Replication/physiology
7.
EMBO J ; 37(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29789390

ABSTRACT

Cytoplasmic lipid droplets are important organelles in nearly every eukaryotic and some prokaryotic cells. Storing and providing energy is their main function, but they do not work in isolation. They respond to stimuli initiated either on the cell surface or in the cytoplasm as conditions change. Cellular stresses such as starvation and invasion are internal insults that evoke changes in droplet metabolism and dynamics. This review will first outline lipid droplet assembly and then discuss how droplets respond to stress and in particular nutrient starvation. Finally, the role of droplets in viral and microbial invasion will be presented, where an unresolved issue is whether changes in droplet abundance promote the invader, defend the host, to try to do both. The challenges of stress and infection are often accompanied by changes in physical contacts between droplets and other organelles. How these changes may result in improving cellular physiology, an ongoing focus in the field, is discussed.


Subject(s)
Bacterial Infections/metabolism , Cytoplasm/metabolism , Lipid Droplets/metabolism , Stress, Physiological , Virus Diseases/metabolism , Animals , Bacterial Infections/pathology , Cytoplasm/microbiology , Cytoplasm/pathology , Cytoplasm/virology , Humans , Lipid Droplets/microbiology , Lipid Droplets/pathology , Lipid Droplets/virology , Virus Diseases/pathology
8.
PLoS Pathog ; 16(6): e1008554, 2020 06.
Article in English | MEDLINE | ID: mdl-32542055

ABSTRACT

Lipid droplets are essential cellular organelles for storage of fatty acids and triglycerides. The hepatitis C virus (HCV) translocates several of its proteins onto their surface and uses them for production of infectious progeny. We recently reported that the lipid droplet-associated α/ß hydrolase domain-containing protein 5 (ABHD5/CGI-58) participates in HCV assembly by mobilizing lipid droplet-associated lipids. However, ABHD5 itself has no lipase activity and it remained unclear how ABHD5 mediates lipolysis critical for HCV assembly. Here, we identify adipose triglyceride lipase (ATGL) as ABHD5 effector and new host factor involved in the hepatic lipid droplet degradation as well as in HCV and lipoprotein morphogenesis. Modulation of ATGL protein expression and lipase activity controlled lipid droplet lipolysis and virus production. ABHD4 is a paralog of ABHD5 unable to activate ATGL or support HCV assembly and lipid droplet lipolysis. Grafting ABHD5 residues critical for activation of ATGL onto ABHD4 restored the interaction between lipase and co-lipase and bestowed the pro-viral and lipolytic functions onto the engineered protein. Congruently, mutation of the predicted ABHD5 protein interface to ATGL ablated ABHD5 functions in lipid droplet lipolysis and HCV assembly. Interestingly, minor alleles of ABHD5 and ATGL associated with neutral lipid storage diseases in human, are also impaired in lipid droplet lipolysis and their pro-viral functions. Collectively, these results show that ABHD5 cooperates with ATGL to mobilize triglycerides for HCV infectious virus production. Moreover, viral manipulation of lipid droplet homeostasis via the ABHD5-ATGL axis, akin to natural genetic variation in these proteins, emerges as a possible mechanism by which chronic HCV infection causes liver steatosis.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Hepacivirus/physiology , Lipase/metabolism , Lipid Droplets/metabolism , Lipolysis , Virus Assembly/physiology , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Humans , Lipase/genetics , Lipid Droplets/virology , Triglycerides/genetics , Triglycerides/metabolism
9.
J Cell Sci ; 132(1)2019 01 09.
Article in English | MEDLINE | ID: mdl-30559250

ABSTRACT

In hepatocytes, PLIN2 is the major protein coating lipid droplets (LDs), an organelle the hepatitis C virus (HCV) hijacks for virion morphogenesis. We investigated the consequences of PLIN2 deficiency on LDs and on HCV infection. Knockdown of PLIN2 did not affect LD homeostasis, likely due to compensation by PLIN3, but severely impaired HCV particle production. PLIN2-knockdown cells had slightly larger LDs with altered protein composition, enhanced local lipase activity and higher ß-oxidation capacity. Electron micrographs showed that, after PLIN2 knockdown, LDs and HCV-induced vesicular structures were tightly surrounded by ER-derived double-membrane sacs. Strikingly, the LD access for HCV core and NS5A proteins was restricted in PLIN2-deficient cells, which correlated with reduced formation of intracellular HCV particles that were less infectious and of higher density, indicating defects in maturation. PLIN2 depletion also reduced protein levels and secretion of ApoE due to lysosomal degradation, but did not affect the density of ApoE-containing lipoproteins. However, ApoE overexpression in PLIN2-deficient cells did not restore HCV spreading. Thus, PLIN2 expression is required for trafficking of core and NS5A proteins to LDs, and for formation of functional low-density HCV particles prior to ApoE incorporation.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Hepacivirus/pathogenicity , Hepatitis C/virology , Hepatocytes/virology , Lipid Droplets/virology , Lipoproteins/metabolism , Perilipin-2/metabolism , Virion/physiology , HEK293 Cells , Hepatitis C/metabolism , Hepatocytes/metabolism , Humans , Lipid Droplets/metabolism , Perilipin-2/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
10.
J Virol ; 94(15)2020 07 16.
Article in English | MEDLINE | ID: mdl-32461314

ABSTRACT

Many RNA viruses replicate in cytoplasmic compartments (virus factories or viroplasms) composed of viral and cellular proteins, but the mechanisms required for their formation remain largely unknown. Rotavirus (RV) replication in viroplasms requires interactions between virus nonstructural proteins NSP2 and NSP5, which are associated with components of lipid droplets (LDs). We previously identified two forms of NSP2 in RV-infected cells, a cytoplasmically dispersed form (dNSP2) and a viroplasm-specific form (vNSP2), which interact with hypophosphorylated and hyperphosphorylated NSP5, respectively, indicating that a coordinated phosphorylation cascade controls viroplasm assembly. The cellular kinase CK1α phosphorylates NSP2 on serine 313, triggering the localization of vNSP2 to sites of viroplasm assembly and its association with hyperphosphorylated NSP5. Using reverse genetics, we generated a rotavirus with a phosphomimetic NSP2 (S313D) mutation to directly evaluate the role of CK1α NSP2 phosphorylation in viroplasm formation. Recombinant rotavirus NSP2 S313D (rRV NSP2 S313D) is significantly delayed in viroplasm formation and in virus replication and interferes with wild-type RV replication in coinfection. Taking advantage of the delay in viroplasm formation, the NSP2 phosphomimetic mutant was used as a tool to observe very early events in viroplasm assembly. We show that (i) viroplasm assembly correlates with NSP5 hyperphosphorylation and (ii) vNSP2 S313D colocalizes with RV-induced LDs without NSP5, suggesting that vNSP2 phospho-S313 is sufficient for interacting with LDs and may be the virus factor required for RV-induced LD formation. Further studies with the rRV NSP2 S313D virus are expected to reveal new aspects of viroplasm and LD initiation and assembly.IMPORTANCE Reverse genetics was used to generate a recombinant rotavirus with a single phosphomimetic mutation in nonstructural protein 2 (NSP2 S313D) that exhibits delayed viroplasm formation, delayed replication, and an interfering phenotype during coinfection with wild-type rotavirus, indicating the importance of this amino acid during virus replication. Exploiting the delay in viroplasm assembly, we found that viroplasm-associated NSP2 colocalizes with rotavirus-induced lipid droplets prior to the accumulation of other rotavirus proteins that are required for viroplasm formation and that NSP5 hyperphosphorylation is required for viroplasm assembly. These data suggest that NSP2 phospho-S313 is sufficient for interaction with lipid droplets and may be the virus factor that induces lipid droplet biogenesis in rotavirus-infected cells. Lipid droplets are cellular organelles critical for the replication of many viral and bacterial pathogens, and thus, understanding the mechanism of NSP2-mediated viroplasm/lipid droplet initiation and interaction will lead to new insights into this important host-pathogen interaction.


Subject(s)
Lipid Droplets/metabolism , Lipid Droplets/virology , RNA-Binding Proteins/metabolism , Rotavirus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , Animals , Cell Line , Cricetinae , Phosphorylation , RNA-Binding Proteins/genetics , Viral Nonstructural Proteins/genetics
11.
Int J Mol Sci ; 22(11)2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34198853

ABSTRACT

In nonalcoholic steatohepatitis animal models, an increased lipid droplet size in hepatocytes is associated with fibrogenesis. Hepatocytes with large droplet (Ld-MaS) or small droplet (Sd-MaS) macrovesicular steatosis may coexist in the human liver, but the factors associated with the predominance of one type over the other, including hepatic fibrogenic capacity, are unknown. In pre-ischemic liver biopsies from 225 consecutive liver transplant donors, we retrospectively counted hepatocytes with Ld-MaS and Sd-MaS and defined the predominant type of steatosis as involving ≥50% of steatotic hepatocytes. We analyzed a donor Patatin-like phospholipase domain-containing protein 3 (PNPLA3) rs738409 polymorphism, hepatic expression of proteins involved in lipid metabolism by RT-PCR, hepatic stellate cell (HSC) activation by α-SMA immunohistochemistry and, one year after transplantation, histological progression of fibrosis due to Hepatitis C Virus (HCV) recurrence. Seventy-four livers had no steatosis, and there were 98 and 53 with predominant Ld-MaS and Sd-MaS, respectively. In linear regression models, adjusted for many donor variables, the percentage of steatotic hepatocytes affected by Ld-MaS was inversely associated with hepatic expression of Insulin Induced Gene 1 (INSIG-1) and Niemann-Pick C1-Like 1 gene (NPC1L1) and directly with donor PNPLA3 variant M, HSC activation and progression of post-transplant fibrosis. In humans, Ld-MaS formation by hepatocytes is associated with abnormal PNPLA3-mediated lipolysis, downregulation of both the intracellular cholesterol sensor and cholesterol reabsorption from bile and increased hepatic fibrogenesis.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Lipase/genetics , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Adult , Aged , Female , Gene Expression Regulation/genetics , Hepacivirus/genetics , Hepatocytes/virology , Humans , Lipid Droplets/metabolism , Lipid Droplets/pathology , Lipid Droplets/virology , Liver/metabolism , Liver/pathology , Liver/virology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/virology , Polymorphism, Single Nucleotide/genetics , Retrospective Studies
12.
J Virol ; 93(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30728260

ABSTRACT

Retinoid (vitamin A) is an essential diet constituent that governs a broad range of biological processes. Its biologically active metabolite, all-trans retinoic acid (ATRA), exhibits a potent antiviral property by enhancing both innate and adaptive antiviral immunity against a variety of viral pathogens, such as, but not limited to, HIV, respiratory syncytial virus (RSV), herpes simplex virus (HSV), and measles. Even though the hepatocyte is highly enriched with retinoid and its metabolite ATRA, it supports the establishment of efficient hepatitis C virus (HCV) replication. Here, we demonstrate the hepatocyte-specific cell-intrinsic mechanism by which ATRA exerts either a proviral or antiviral effect, depending on how it engages cellular retinoic acid binding proteins (CRABPs). We found that the engagement of CRABP1 by ATRA potently supported viral infection by promoting the accumulation of lipid droplets (LDs), which robustly enhanced the formation of a replication complex on the LD-associated endoplasmic reticulum (ER) membrane. In contrast, ATRA binding to CRABP2 potently inhibited HCV via suppression of LD accumulation. However, this antiviral effect of CRABP2 was abrogated due to the functional and quantitative predominance of CRABP1 in the hepatocytes. In summary, our study demonstrates that CRABPs serve as an on-off switch that modulates the efficiency of the HCV life cycle and elucidates how HCV evades the antiviral properties of ATRA via the exploitation of CRABP1 functionality.IMPORTANCE ATRA, a biologically active metabolite of vitamin A, exerts pleiotropic biological effects, including the activation of both innate and adaptive immunity, thereby serving as a potent antimicrobial compound against numerous viral pathogens. Despite the enrichment of hepatocytes with vitamin A, HCV still establishes an efficient viral life cycle. Here, we discovered that the hepatocellular response to ATRA creates either a proviral or an antiviral environment depending on its engagement with CRABP1 or -2, respectively. CRABP1 supports the robust replication of HCV, while CRABP2 potently inhibits the efficiency of viral replication. Our biochemical, genetic, and microscopic analyses reveal that the pro- and antiviral effects of CRABPs are mediated by modulation of LD abundance, where HCV establishes the platform for viral replication and assembly on the LD-associated ER membrane. This study uncovered a cell-intrinsic mechanism by which HCV exploits the proviral function of CRABP1 to establish an efficient viral life cycle.


Subject(s)
Hepacivirus/metabolism , Hepatitis C/metabolism , Lipid Droplets/metabolism , Receptors, Retinoic Acid/metabolism , Antiviral Agents/pharmacology , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Hepatitis C/pathology , Humans , Lipid Droplets/virology , Tretinoin/pharmacology
13.
Cell Microbiol ; 21(10): e13086, 2019 10.
Article in English | MEDLINE | ID: mdl-31290220

ABSTRACT

The cellular lipid pool plays a central role in hepatitis C virus (HCV) life cycle, from establishing infection to virus propagation. Here, we show that a liver abundant long noncoding RNA, highly upregulated in liver carcinoma (HULC), is upregulated during HCV infection and manipulates the lipid pool to favour virus life cycle. Interestingly, HULC was found to be crucial for the increase in number of lipid droplets in infected cells. This effect was attributed to the role of HULC in lipid biogenesis. Further, we demonstrated that HULC knockdown decreases the association of HCV-core protein with lipid droplets. This exhibited a direct consequence on the release of HCV particles. The role of HULC in HCV-particle release was further substantiated by additional knockdown and mutation experiments. Additionally, we found that increased level of HULC in HCV-infected cells was a result of Retinoid X Receptor Alpha (RXRA)-mediated transcription, which seemed to be aided by HCV-core protein. Taken together, the results identify a distinct role of long noncoding RNA HULC in lipid dynamics during HCV infection, which provides new insights into the complex process of HCV propagation and pathogenesis.


Subject(s)
Hepacivirus/physiology , Lipid Droplets/metabolism , Liver/metabolism , Liver/virology , RNA, Long Noncoding/metabolism , Viral Core Proteins/metabolism , Virion/metabolism , Cell Line, Tumor , Cell Survival/genetics , Gene Knockdown Techniques , Humans , Lipid Droplets/virology , Lipid Metabolism/genetics , Liver/pathology , RNA, Long Noncoding/genetics , Retinoid X Receptor alpha/metabolism , Viral Core Proteins/genetics
14.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114346

ABSTRACT

Hijacking and manipulation of host cell biosynthetic pathways by human enveloped viruses are essential for the viral lifecycle. Flaviviridae members, including hepatitis C, dengue and Zika viruses, extensively manipulate host lipid metabolism, underlining the importance of lipid droplets (LDs) in viral infection. LDs are dynamic cytoplasmic organelles that can act as sequestration platforms for a unique subset of host and viral proteins. Transient recruitment and mobilization of proteins to LDs during viral infection impacts host-cell biological properties, LD functionality and canonical protein functions. Notably, recent studies identified LDs in the nucleus and also identified that LDs are transported extracellularly via an autophagy-mediated mechanism, indicating a novel role for autophagy in Flaviviridae infections. These developments underline an unsuspected diversity and localization of LDs and potential moonlighting functions of LD-associated proteins during infection. This review summarizes recent breakthroughs concerning the LD hijacking activities of hepatitis C, dengue and Zika viruses and potential roles of cytoplasmic, nuclear and extracellular LD-associated viral proteins during infection.


Subject(s)
Flaviviridae/pathogenicity , Lipid Droplets/metabolism , Viral Proteins/metabolism , Animals , Autophagy , Cell Nucleus/metabolism , Dengue Virus/metabolism , Dengue Virus/pathogenicity , Extracellular Space/metabolism , Flaviviridae/metabolism , Hepacivirus/metabolism , Hepacivirus/pathogenicity , Humans , Lipid Droplets/virology , Zika Virus/metabolism , Zika Virus/pathogenicity
15.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 1041-1056, 2018 09.
Article in English | MEDLINE | ID: mdl-29885363

ABSTRACT

The hepatitis C virus (HCV) life cycle is tightly linked to the host cell lipid metabolism with the endoplasmic reticulum-derived membranous web harboring viral RNA replication complexes and lipid droplets as virion assembly sites. To investigate HCV-induced changes in the lipid composition, we performed quantitative shotgun lipidomic studies of whole cell extracts and subcellular compartments. Our results indicate that HCV infection reduces the ratio of neutral to membrane lipids. While the amount of neutral lipids and lipid droplet morphology were unchanged, membrane lipids, especially cholesterol and phospholipids, accumulated in the microsomal fraction in HCV-infected cells. In addition, HCV-infected cells had a higher relative abundance of phosphatidylcholines and triglycerides with longer fatty acyl chains and a strikingly increased utilization of C18 fatty acids, most prominently oleic acid (FA [18:1]). Accordingly, depletion of fatty acid elongases and desaturases impaired HCV replication. Moreover, the analysis of free fatty acids revealed increased levels of polyunsaturated fatty acids (PUFAs) caused by HCV infection. Interestingly, inhibition of the PUFA synthesis pathway via knockdown of the rate-limiting Δ6-desaturase enzyme or by treatment with a high dose of a small-molecule inhibitor impaired viral progeny production, indicating that elevated PUFAs are needed for virion morphogenesis. In contrast, pretreatment with low inhibitor concentrations promoted HCV translation and/or early RNA replication. Taken together our results demonstrate the complex remodeling of the host cell lipid metabolism induced by HCV to enhance both virus replication and progeny production.


Subject(s)
Hepacivirus/metabolism , Hepatocytes/metabolism , Host-Pathogen Interactions , Lipid Metabolism/genetics , Metabolome , Virion/metabolism , Virus Replication/physiology , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/genetics , Acetyltransferases/metabolism , Cell Line, Tumor , Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Fatty Acid Desaturases/antagonists & inhibitors , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Fatty Acid Elongases , Fatty Acids, Unsaturated/metabolism , Gene Expression Regulation , Hepacivirus/growth & development , Hepatocytes/chemistry , Hepatocytes/virology , Humans , Lipid Droplets/metabolism , Lipid Droplets/virology , Microsomes/metabolism , Microsomes/virology , Oleic Acid/metabolism , Phosphatidylcholines/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/biosynthesis , RNA, Viral/genetics , Triglycerides/metabolism , Virion/growth & development , Virus Assembly/physiology
16.
IUBMB Life ; 70(1): 41-49, 2018 01.
Article in English | MEDLINE | ID: mdl-29281185

ABSTRACT

Hepatitis C virus (HCV) has infected over 170 million people world-wide. This infection causes severe liver damage that can progress to hepatocellular carcinoma leading to death of the infected patients. Development of a cell culture model system for the study of HCV infection in the recent past has helped the researchers world-wide to understand the biology of this virus. Studies over the past decade have revealed the tricks played by the virus to sustain itself, for as long as 40 years, in the host setup without being eliminated by the immune system. Today we understand that the host organelles and different cellular proteins are affected during HCV infection. This cytoplasmic virus has all the cellular organelles at its disposal to successfully replicate, from ribosomes and intracellular membranous structures to the nucleus. It modulates these organelles at both the structural and the functional levels. The vast knowledge about the viral genome and viral proteins has also helped in the development of drugs against the virus. Despite the achieved success rate to cure the infected patients, we struggle to eliminate the cases of recurrence and the non-responders. Such cases might emerge owing to the property of the viral genome to accumulate mutations during its succeeding replication cycles which favours its survival. The current situation calls an urgent need for alternate therapeutic strategies to counter this major problem of human health. © 2017 IUBMB Life, 70(1):41-49, 2018.


Subject(s)
Carcinoma, Hepatocellular/virology , Hepacivirus/pathogenicity , Hepatitis C, Chronic/virology , Hepatocytes/virology , Immune Evasion , Liver Neoplasms/virology , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/prevention & control , Cell Nucleus/immunology , Cell Nucleus/virology , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/virology , Gene Expression Regulation , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Hepatocytes/immunology , Humans , Lipid Droplets/immunology , Lipid Droplets/virology , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/immunology , Liver Neoplasms/etiology , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , RNA, Viral/biosynthesis , RNA, Viral/genetics , Ribosomes/immunology , Ribosomes/virology , Signal Transduction , Viral Proteins/genetics , Viral Proteins/immunology , Virus Replication/drug effects
17.
Appl Environ Microbiol ; 84(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29625986

ABSTRACT

Infectious diseases caused by enveloped viruses, such as influenza, severe acute respiratory syndrome (SARS), and Middle East respiratory syndrome (MERS), cause thousands of deaths and billions of dollars of economic losses per year. Studies have found a relationship among temperature, humidity, and influenza virus incidence, transmission, or survival; however, there are contradictory claims about whether absolute humidity (AH) or relative humidity (RH) is most important in mediating virus infectivity. Using the enveloped bacteriophage Phi6, which has been suggested as a surrogate for influenza viruses and coronaviruses, we designed a study to discern whether AH, RH, or temperature is a better predictor of virus survival in droplets. Our results show that Phi6 survived best at high (>85%) and low (<60%) RHs, with a significant decrease in infectivity at mid-range RHs (∼60 to 85%). At an AH of less than 22 g · m-3, the loss in infectivity was less than 2 orders of magnitude; however, when the AH was greater than 22 g · m-3, the loss in infectivity was typically greater than 6 orders of magnitude. At a fixed RH of 75%, infectivity was very sensitive to temperature, decreasing two orders of magnitude between 19°C and 25°C. We used random forest modeling to identify the best environmental predictors for modulating virus infectivity. The model explained 83% of variation in Phi6 infectivity and suggested that RH is the most important factor in controlling virus infectivity in droplets. This research provides novel information about the complex interplay between temperature, humidity, and the survival of viruses in droplets.IMPORTANCE Enveloped viruses are responsible for a number of infectious diseases resulting in thousands of deaths and billions of dollars of economic losses per year in the United States. There has been a lively debate in the literature over whether absolute humidity (AH) or relative humidity (RH) modulates virus infectivity. We designed a controlled study and used advanced statistical modeling techniques specifically to address this question. By providing an improved understanding of the relationship between environmental conditions and virus infectivity, our work will ultimately lead to improved strategies for predicting and controlling disease transmission.


Subject(s)
Bacteriophages/physiology , Humidity , Lipid Droplets/virology , Temperature , Virus Inactivation , Virus Physiological Phenomena
18.
Cell Microbiol ; 19(1)2017 01.
Article in English | MEDLINE | ID: mdl-27794207

ABSTRACT

Lipid droplets were long considered to be simple storage structures, but they have recently been shown to be dynamic organelles involved in diverse biological processes, including emerging roles in innate immunity. Various intracellular pathogens, including viruses, bacteria, and parasites, specifically target host lipid droplets during their life cycle. Viruses such as hepatitis C, dengue, and rotaviruses use lipid droplets as platforms for assembly. Bacteria, such as mycobacteria and Chlamydia, and parasites, such as trypanosomes, use host lipid droplets for nutritional purposes. The possible use of lipid droplets by intracellular pathogens, as part of an anti-immunity strategy, is an intriguing question meriting further investigation in the near future.


Subject(s)
Bacteria/metabolism , Lipid Droplets/immunology , Trypanosoma/metabolism , Virus Assembly , Animals , Humans , Lipid Droplets/microbiology , Lipid Droplets/parasitology , Lipid Droplets/virology , Lipid Metabolism
19.
Traffic ; 16(9): 962-77, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26031340

ABSTRACT

Dengue viruses cause the most important human viral disease transmitted by mosquitoes. In recent years, a great deal has been learned about molecular details of dengue virus genome replication; however, little is known about genome encapsidation and the functions of the viral capsid protein. During infection, dengue virus capsid progressively accumulates around lipid droplets (LDs) by an unknown mechanism. Here, we examined the process by which the viral capsid is transported from the endoplasmic reticulum (ER) membrane, where the protein is synthesized, to LDs. Using different methods of intervention, we found that the GBF1-Arf1/Arf4-COPI pathway is necessary for capsid transport to LDs, while the process is independent of both COPII components and Golgi integrity. The transport was sensitive to Brefeldin A, while a drug resistant form of GBF1 was sufficient to restore capsid subcellular distribution in infected cells. The mechanism by which LDs gain or lose proteins is still an open question. Our results support a model in which the virus uses a non-canonical function of the COPI system for capsid accumulation on LDs, providing new ideas for antiviral strategies.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Capsid Proteins/metabolism , Coat Protein Complex I/metabolism , Dengue Virus/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Lipid Droplets/metabolism , Cell Line, Tumor , Dengue Virus/pathogenicity , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Humans , Lipid Droplets/virology , Protein Transport
20.
J Lipid Res ; 58(2): 420-432, 2017 02.
Article in English | MEDLINE | ID: mdl-27941027

ABSTRACT

Hepatitis C virus (HCV) is an enveloped RNA virus responsible for 170 million cases of viral hepatitis worldwide. Over 50% of chronically infected HCV patients develop hepatic steatosis, and steatosis can be induced by expression of HCV core protein (core) alone. Additionally, core must associate with cytoplasmic lipid droplets (LDs) for steatosis development and viral particle assembly. Due to the importance of the LD as a key component of hepatic lipid storage and as a platform for HCV particle assembly, it seems this dynamic subcellular organelle is a gatekeeper in the pathogenesis of viral hepatitis. Here, we hypothesized that core requires the host LD scaffold protein, perilipin (PLIN)3, to induce hepatic steatosis. To test our hypothesis in vivo, we have studied core-induced hepatic steatosis in the absence or presence of antisense oligonucleotide-mediated knockdown of PLIN3. PLIN3 knockdown blunted HCV core-induced steatosis in transgenic mice fed either chow or a moderate fat diet. Collectively, our studies demonstrate that the LD scaffold protein, PLIN3, is essential for HCV core-induced hepatic steatosis and provide new insights into the pathogenesis of HCV.


Subject(s)
Fatty Liver/genetics , Hepatitis C/metabolism , Liver/metabolism , Perilipin-3/genetics , Animals , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Knockdown Techniques , Genotype , Hepacivirus , Hepatitis C/genetics , Hepatitis C/pathology , Hepatitis C/virology , Humans , Lipid Droplets/metabolism , Lipid Droplets/pathology , Lipid Droplets/virology , Lipid Metabolism/genetics , Liver/pathology , Liver/virology , Mice , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Perilipin-3/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL