Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cell ; 185(21): 3980-3991.e18, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36182704

ABSTRACT

Simian arteriviruses are endemic in some African primates and can cause fatal hemorrhagic fevers when they cross into primate hosts of new species. We find that CD163 acts as an intracellular receptor for simian hemorrhagic fever virus (SHFV; a simian arterivirus), a rare mode of virus entry that is shared with other hemorrhagic fever-causing viruses (e.g., Ebola and Lassa viruses). Further, SHFV enters and replicates in human monocytes, indicating full functionality of all of the human cellular proteins required for viral replication. Thus, simian arteriviruses in nature may not require major adaptations to the human host. Given that at least three distinct simian arteriviruses have caused fatal infections in captive macaques after host-switching, and that humans are immunologically naive to this family of viruses, development of serology tests for human surveillance should be a priority.


Subject(s)
Arterivirus , Hemorrhagic Fevers, Viral , Animals , Arterivirus/physiology , Hemorrhagic Fevers, Viral/veterinary , Hemorrhagic Fevers, Viral/virology , Humans , Macaca , Primates , Viral Zoonoses , Virus Internalization , Virus Replication
2.
Viruses ; 13(4)2021 04 07.
Article in English | MEDLINE | ID: mdl-33917085

ABSTRACT

Simian hemorrhagic fever virus (SHFV) causes acute, lethal disease in macaques. We developed a single-plasmid cDNA-launch infectious clone of SHFV (rSHFV) and modified the clone to rescue an enhanced green fluorescent protein-expressing rSHFV-eGFP that can be used for rapid and quantitative detection of infection. SHFV has a narrow cell tropism in vitro, with only the grivet MA-104 cell line and a few other grivet cell lines being susceptible to virion entry and permissive to infection. Using rSHFV-eGFP, we demonstrate that one cricetid rodent cell line and three ape cell lines also fully support SHFV replication, whereas 55 human cell lines, 11 bat cell lines, and three rodent cells do not. Interestingly, some human and other mammalian cell lines apparently resistant to SHFV infection are permissive after transfection with the rSHFV-eGFP cDNA-launch plasmid. To further demonstrate the investigative potential of the infectious clone system, we introduced stop codons into eight viral open reading frames (ORFs). This approach suggested that at least one ORF, ORF 2b', is dispensable for SHFV in vitro replication. Our proof-of-principle experiments indicated that rSHFV-eGFP is a useful tool for illuminating the understudied molecular biology of SHFV.


Subject(s)
Arterivirus/genetics , DNA, Complementary/genetics , Green Fluorescent Proteins/genetics , Open Reading Frames , RNA, Viral/genetics , Recombination, Genetic , Virus Replication/genetics , Animals , Arterivirus/physiology , Cell Line , Chiroptera , Hominidae , Humans , Plasmids/genetics , Proof of Concept Study , Rodentia
3.
Virology ; 517: 148-156, 2018 04.
Article in English | MEDLINE | ID: mdl-29249267

ABSTRACT

Cyclophilin A (CypA) is an important host factor in the replication of a variety of RNA viruses. Also the replication of several nidoviruses was reported to depend on CypA, although possibly not to the same extent. These prior studies are difficult to compare, since different nidoviruses, cell lines and experimental set-ups were used. Here, we investigated the CypA dependence of three distantly related nidoviruses that can all replicate in Huh7 cells: the arterivirus equine arteritis virus (EAV), the alphacoronavirus human coronavirus 229E (HCoV-229E), and the betacoronavirus Middle East respiratory syndrome coronavirus (MERS-CoV). The replication of these viruses was compared in the same parental Huh7 cells and in CypA-knockout Huh7 cells generated using CRISPR/Cas9-technology. CypA depletion reduced EAV yields by ~ 3-log, whereas MERS-CoV progeny titers were modestly reduced (3-fold) and HCoV-229E replication was unchanged. This study reveals that the replication of nidoviruses can differ strikingly in its dependence on cellular CypA.


Subject(s)
Arterivirus/physiology , Coronavirus/physiology , Cyclophilin A/metabolism , Virus Cultivation , Virus Replication/physiology , Animals , Cell Line , Cricetinae , Humans
4.
J Virol ; 91(4)2017 02 15.
Article in English | MEDLINE | ID: mdl-27974564

ABSTRACT

Simian arteriviruses are a diverse clade of viruses infecting captive and wild nonhuman primates. We recently reported that Kibale red colobus virus 1 (KRCV-1) causes a mild and self-limiting disease in experimentally infected crab-eating macaques, while simian hemorrhagic fever virus (SHFV) causes lethal viral hemorrhagic fever. Here we characterize how these viruses evolved during replication in cell culture and in experimentally infected macaques. During passage in cell culture, 68 substitutions that were localized in open reading frames (ORFs) likely associated with host cell entry and exit became fixed in the KRCV-1 genome. However, we did not detect any strong signatures of selection during replication in macaques. We uncovered patterns of evolution that were distinct from those observed in surveys of wild red colobus monkeys, suggesting that these species may exert different adaptive challenges for KRCV-1. During SHFV infection, we detected signatures of selection on ORF 5a and on a small subset of sites in the genome. Overall, our data suggest that patterns of evolution differ markedly among simian arteriviruses and among host species. IMPORTANCE: Certain RNA viruses can cross species barriers and cause disease in new hosts. Simian arteriviruses are a diverse group of related viruses that infect captive and wild nonhuman primates, with associated disease severity ranging from apparently asymptomatic infections to severe, viral hemorrhagic fevers. We infected nonhuman primate cell cultures and then crab-eating macaques with either simian hemorrhagic fever virus (SHFV) or Kibale red colobus virus 1 (KRCV-1) and assessed within-host viral evolution. We found that KRCV-1 quickly acquired a large number of substitutions in its genome during replication in cell culture but that evolution in macaques was limited. In contrast, we detected selection focused on SHFV ORFs 5a and 5, which encode putative membrane proteins. These patterns suggest that in addition to diverse pathogenic phenotypes, these viruses may also exhibit distinct patterns of within-host evolution both in vitro and in vivo.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/physiology , Biological Evolution , Host-Pathogen Interactions , Monkey Diseases/virology , Animals , Host-Pathogen Interactions/genetics , Macaca fascicularis , Monkey Diseases/genetics , Open Reading Frames , Polymorphism, Single Nucleotide , RNA, Viral , Selection, Genetic , Virus Internalization , Virus Replication
5.
mBio ; 7(6)2016 12 06.
Article in English | MEDLINE | ID: mdl-27923923

ABSTRACT

Infection with nidoviruses like corona- and arteriviruses induces a reticulovesicular network of interconnected endoplasmic reticulum (ER)-derived double-membrane vesicles (DMVs) and other membrane structures. This network is thought to accommodate the viral replication machinery and protect it from innate immune detection. We hypothesized that the innate immune response has tools to counteract the formation of these virus-induced replication organelles in order to inhibit virus replication. Here we have investigated the effect of type I interferon (IFN) treatment on the formation of arterivirus-induced membrane structures. Our approach involved ectopic expression of arterivirus nonstructural proteins nsp2 and nsp3, which induce DMV formation in the absence of other viral triggers of the interferon response, such as replicating viral RNA. Thus, this setup can be used to identify immune effectors that specifically target the (formation of) virus-induced membrane structures. Using large-scale electron microscopy mosaic maps, we found that IFN-ß treatment significantly reduced the formation of the membrane structures. Strikingly, we also observed abundant stretches of double-membrane sheets (a proposed intermediate of DMV formation) in IFN-ß-treated samples, suggesting the disruption of DMV biogenesis. Three interferon-stimulated gene products, two of which have been reported to target the hepatitis C virus replication structures, were tested for their possible involvement, but none of them affected membrane structure formation. Our study reveals the existence of a previously unknown innate immune mechanism that antagonizes the viral hijacking of host membranes. It also provides a solid basis for further research into the poorly understood interactions between the innate immune system and virus-induced replication structures. IMPORTANCE: Viruses with a positive-strand RNA genome establish a membrane-associated replication organelle by hijacking and remodeling intracellular host membranes, a process deemed essential for their efficient replication. It is unknown whether the cellular innate immune system can detect and/or inhibit the formation of these membrane structures, which could be an effective mechanism to delay viral RNA replication. In this study, using an expression system that closely mimics the formation of arterivirus replication structures, we show for the first time that IFN-ß treatment clearly reduces the amount of induced membrane structures. Moreover, drastic morphological changes were observed among the remaining structures, suggesting that their biogenesis was impaired. Follow-up experiments suggested that host cells contain a hitherto unknown innate antiviral mechanism, which targets this common feature of positive-strand RNA virus replication. Our study provides a strong basis for further research into the interaction of the innate immune system with membranous viral replication organelles.


Subject(s)
Arterivirus/immunology , Arterivirus/physiology , Immunity, Innate , Interferon-beta/metabolism , Intracellular Membranes/metabolism , Intracellular Membranes/virology , Virus Replication , Microscopy, Electron, Transmission , Viral Nonstructural Proteins/metabolism
6.
Dev Comp Immunol ; 65: 245-252, 2016 12.
Article in English | MEDLINE | ID: mdl-27473784

ABSTRACT

The aim of this study was to characterize the immune response of dendritic cells derived from monocytes (Mo-DCs) in the porcine peripheral blood following infection with porcine reproductive and respiratory syndrome virus (PRRSV). Viral load assays indicated that PRRSV efficiently infected Mo-DCs but failed to replicate, whereas PRRSV infection of Mo-DCs decreased the expression of SLA-I, SLA-II, CD80 and CD40 compared with those of mock Mo-DCs. Furthermore, we analyzed the cytokine profiles using quantitative RT-PCR and ELISA. Results indicated apparent changes in IL-10 and IL-12 p40 expression but not in IFN-γ and TNF-α among Mo-DCs infected with PRRSV and uninfected Mo-DCs. Additionally, flow cytometry analysis of the altered Mo-DCs together with IL-4 and GM-CSF induction for 7days revealed the typical morphology and phenotype with 91.73% purity before infection with PRRSV. Overall, our data demonstrate that PRRSV impaired the normal antigen presentation of Mo-DCs and led to inadequate adaptive immune response by down-regulating the expression of SLA-I,SLA-II, CD80 and CD40. Enhanced Th2 -type cytokine IL-10 secretion and reduced Th1-type cytokines IL-12p40,IFN-γ and TNF-α secretion results in Th1/Th2 imbalance.


Subject(s)
Adaptive Immunity , Arterivirus Infections/immunology , Arterivirus/physiology , Dendritic Cells/immunology , Monocytes/immunology , Animals , Antigen Presentation , Cell Differentiation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/virology , Histocompatibility Antigens/metabolism , Immune Evasion , Swine , Th1-Th2 Balance , Transcriptome , Viral Load
7.
J Virol ; 90(2): 630-5, 2016 01 15.
Article in English | MEDLINE | ID: mdl-26559828

ABSTRACT

Wild nonhuman primates are immediate sources and long-term reservoirs of human pathogens. However, ethical and technical challenges have hampered the identification of novel blood-borne pathogens in these animals. We recently examined RNA viruses in plasma from wild African monkeys and discovered several novel, highly divergent viruses belonging to the family Arteriviridae. Close relatives of these viruses, including simian hemorrhagic fever virus, have caused sporadic outbreaks of viral hemorrhagic fever in captive macaque monkeys since the 1960s. However, arterivirus infection in wild nonhuman primates had not been described prior to 2011. The arteriviruses recently identified in wild monkeys have high sequence and host species diversity, maintain high viremia, and are prevalent in affected populations. Taken together, these features suggest that the simian arteriviruses may be "preemergent" zoonotic pathogens. If not, this would imply that biological characteristics of RNA viruses thought to facilitate zoonotic transmission may not, by themselves, be sufficient for such transmission to occur.


Subject(s)
Arterivirus Infections/transmission , Arterivirus Infections/veterinary , Arterivirus/physiology , Primate Diseases/transmission , Primate Diseases/virology , Zoonoses/transmission , Zoonoses/virology , Animals , Arterivirus/genetics , Arterivirus Infections/virology , Haplorhini , Humans
8.
DNA Cell Biol ; 34(12): 728-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26398903

ABSTRACT

NLRP3 inflammasome, which is multiprotein complex that induces the maturity and secretion of proinflammatory interleukin-1ß (IL-1ß), takes a bridge between the innate and adaptive immune responses to the invading pathogens. It has been shown that porcine reproductive and respiratory syndrome virus (PRRSV) could activate the NLRP3 inflammasome but induce the host's immunosuppression. This study aims to explore whether PRRSV could encode the component to antagonize the NLRP3 inflammasome. The obtained results showed that PRRSV could induce the expression and secretion of IL-1ß in early infection through the pathway of NLRP3 inflammasome in porcine alveolar macrophages (PAMs), but the levels of pro-IL-1ß mRNA and IL-1ß protein decreased to a degree that was similar to the level of the mock-infected group in later infection. This work also found that PRRSV nonstructural protein (nsp) 11 could inhibit the expression of pro-IL-1ß mRNA induced by lipopolysaccharide (LPS) and the secretion of IL-1ß induced by LPS plus nigericin in PAMs. Furthermore, the mutation studies showed that the endoribonuclease activity was essential for nsp11 to inhibit the secretion of IL-1ß. Therefore, it could be indicated that PRRSV could induce the activation of NLRP3 inflammasome, but the virus encoded nsp11 to inhibit this action.


Subject(s)
Arterivirus/physiology , Carrier Proteins/metabolism , Endoribonucleases/metabolism , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Porcine Reproductive and Respiratory Syndrome/virology , Viral Nonstructural Proteins/metabolism , Animals , Arterivirus/metabolism , Interleukin-1beta/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Messenger/genetics , Swine
9.
J Virol ; 89(15): 8082-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972539

ABSTRACT

Simian hemorrhagic fever (SHF) is lethal for macaques. Based on clinical presentation and serological diagnosis, all reported SHF outbreaks were thought to be caused by different strains of the same virus, simian hemorrhagic fever virus (SHFV; Arteriviridae). Here we show that the SHF outbreaks in Sukhumi in 1964 and in Alamogordo in 1989 were caused not by SHFV but by two novel divergent arteriviruses. Our results indicate that multiple divergent simian arteriviruses can cause SHF.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/isolation & purification , Hemorrhagic Fevers, Viral/veterinary , Macaca/virology , Primate Diseases/virology , Amino Acid Sequence , Animals , Arterivirus/classification , Arterivirus/genetics , Arterivirus/physiology , Arterivirus Infections/history , Arterivirus Infections/virology , Evolution, Molecular , Hemorrhagic Fevers, Viral/history , Hemorrhagic Fevers, Viral/virology , History, 20th Century , Humans , Molecular Sequence Data , Phylogeny , Primate Diseases/history , Sequence Homology, Amino Acid , Viral Proteins/chemistry , Viral Proteins/genetics
10.
J Virol ; 89(1): 844-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25355889

ABSTRACT

UNLABELLED: Simian hemorrhagic fever virus (SHFV) causes a severe and almost uniformly fatal viral hemorrhagic fever in Asian macaques but is thought to be nonpathogenic for humans. To date, the SHFV life cycle is almost completely uncharacterized on the molecular level. Here, we describe the first steps of the SHFV life cycle. Our experiments indicate that SHFV enters target cells by low-pH-dependent endocytosis. Dynamin inhibitors, chlorpromazine, methyl-ß-cyclodextrin, chloroquine, and concanamycin A dramatically reduced SHFV entry efficiency, whereas the macropinocytosis inhibitors EIPA, blebbistatin, and wortmannin and the caveolin-mediated endocytosis inhibitors nystatin and filipin III had no effect. Furthermore, overexpression and knockout study and electron microscopy results indicate that SHFV entry occurs by a dynamin-dependent clathrin-mediated endocytosis-like pathway. Experiments utilizing latrunculin B, cytochalasin B, and cytochalasin D indicate that SHFV does not hijack the actin polymerization pathway. Treatment of target cells with proteases (proteinase K, papain, α-chymotrypsin, and trypsin) abrogated entry, indicating that the SHFV cell surface receptor is a protein. Phospholipases A2 and D had no effect on SHFV entry. Finally, treatment of cells with antibodies targeting CD163, a cell surface molecule identified as an entry factor for the SHFV-related porcine reproductive and respiratory syndrome virus, diminished SHFV replication, identifying CD163 as an important SHFV entry component. IMPORTANCE: Simian hemorrhagic fever virus (SHFV) causes highly lethal disease in Asian macaques resembling human illness caused by Ebola or Lassa virus. However, little is known about SHFV's ecology and molecular biology and the mechanism by which it causes disease. The results of this study shed light on how SHFV enters its target cells. Using electron microscopy and inhibitors for various cellular pathways, we demonstrate that SHFV invades cells by low-pH-dependent, actin-independent endocytosis, likely with the help of a cellular surface protein.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Arterivirus/physiology , Endocytosis , Host-Pathogen Interactions , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Virus Internalization , Animals , Cell Line , Chlorocebus aethiops
11.
Virus Res ; 202: 112-9, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25455336

ABSTRACT

The simian hemorrhagic fever virus (SHFV) genome differs from those of other members of the family Arteriviridae in encoding three papain-like one proteases (PLP1α, PLP1ß and PLP1γ) at the 5' end and two adjacent sets of four minor structural proteins at the 3' end. The catalytic Cys and His residues and cleavage sites for each of the SHFV PLP1s were predicted and their functionality was tested in in vitro transcription/translation reactions done with wildtype or mutant polyprotein constructs. Mass spectrometry analyses of selected autoproteolytic products confirmed cleavage site locations. The catalytic Cys of PLP1α is unusual in being adjacent to an Ala instead of a Typ. PLP1γ cleaves at both downstream and upstream sites. Intermediate precursor and alternative cleavage products were detected in the in vitro transcription/translation reactions but only the three mature nsp1 proteins were detected in SHFV-infected MA104 cell lysates with SHFV nsp1 protein-specific antibodies. The duplicated sets of SHFV minor structural proteins were predicted to be functionally redundant. A stable, full-length, infectious SHFV-LVR cDNA clone was constructed and a set of mutant infectious clones was generated each with the start codon of one of the minor structural proteins mutated. All eight of the minor structural proteins were found to be required for production of infectious extracellular virus. SHFV causes a fatal hemorrhagic fever in macaques but asymptomatic, persistent infections in natural hosts such as baboons. SHFV infections were compared in macrophages and myeloid dendritic cells from baboons and macaques. Virus yields were higher from macaque cells than from baboon cells. Macrophage cultures from the two types of animals differed dramatically in the percentage of cells infected. In contrast, similar percentages of myeloid dendritic cells were infected but virus replication was efficient in the macaque cells but inefficient in the baboon cells. SHFV infection induced the production of pro-inflammatory cytokines, including IL-1ß, IL-6, IL-12/23(p40), TNF-α and MIP-1α, in macaque cells but not baboon cells.


Subject(s)
Arterivirus/physiology , Papain/metabolism , Viral Structural Proteins/metabolism , Virus Replication , Animals , Arterivirus/enzymology , Arterivirus/genetics , Biomedical Research/trends , Coronavirus Papain-Like Proteases , Cytokines/metabolism , Dendritic Cells/virology , Macaca , Macrophages/virology , Papain/genetics , Papio , Proteolysis , Viral Load , Viral Structural Proteins/genetics
12.
Virus Res ; 194: 100-9, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25262851

ABSTRACT

Arteriviruses infect immune cells and may cause persistence in infected hosts. Inefficient induction of pro-inflammatory cytokines and type I IFNs are observed during infection of this group of viruses, suggesting that they may have evolved to escape the host immune surveillance for efficient survival. Recent studies have identified viral proteins regulating the innate immune signaling, and among these, nsp1 (nonstructural protein 1) is the most potent IFN antagonist. For porcine reproductive and respiratory syndrome virus (PRRSV), individual subunits (nsp1α and nsp1ß) of nsp1 suppress type I IFN production. In particular, PRRSV-nsp1α degrades CREB (cyclic AMP responsive element binding)-binding protein (CBP), a key component of the IFN enhanceosome, whereas PRRSV-nsp1ß degrades karyopherin-α1 which is known to mediate the nuclear import of ISGF3 (interferon-stimulated gene factor 3). All individual subunits of nsp1 of PRRSV, equine arteritis virus (EAV), lactate dehydrogenase-elevating virus (LDV), and simian hemorrhagic fever virus (SHFV) appear to contain IFN suppressive activities. As with PRRSV-nsp1α, CBP degradation is evident by LDV-nsp1α and partly by SHFV-nsp1γ. This review summarizes the biogenesis and the role of individual subunits of nsp1 of arteriviruses for innate immune modulation.


Subject(s)
Arterivirus/immunology , Arterivirus/physiology , Host-Pathogen Interactions , Immune Evasion , Immunity, Innate , Interferon Type I/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism
14.
Virology ; 462-463: 351-62, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25036340

ABSTRACT

The simian hemorrhagic fever virus (SHFV) genome differs from those of other members of the family Arterivirus in encoding two adjacent sets of four minor structural protein open reading frames (ORFs). A stable, full-length, infectious SHFV-LVR cDNA clone was constructed. Virus produced from this clone had replication characteristics similar to those of the parental virus. A subgenomic mRNA was identified for the SHFV ORF previously identified as 2b. As an initial means of analyzing the functional relevance of each of the SHFV minor structural proteins, a set of mutant infectious clones was generated, each with the start codon of one minor structural protein ORF mutated. Different phenotypes were observed for each ortholog of the pairs of minor glycoproteins and all of the eight minor structural proteins were required for the production of infectious extracellular virus indicating that the duplicated sets of SHFV minor structural proteins are not functionally redundant.


Subject(s)
Arterivirus/physiology , Viral Structural Proteins/metabolism , Virus Replication , Animals , Arterivirus/genetics , Cell Line , Chlorocebus aethiops , Codon, Initiator/genetics , Mutation , Viral Structural Proteins/genetics
15.
J Gen Virol ; 94(Pt 10): 2141-2163, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939974

ABSTRACT

Arteriviruses are positive-stranded RNA viruses that infect mammals. They can cause persistent or asymptomatic infections, but also acute disease associated with a respiratory syndrome, abortion or lethal haemorrhagic fever. During the past two decades, porcine reproductive and respiratory syndrome virus (PRRSV) and, to a lesser extent, equine arteritis virus (EAV) have attracted attention as veterinary pathogens with significant economic impact. Particularly noteworthy were the 'porcine high fever disease' outbreaks in South-East Asia and the emergence of new virulent PRRSV strains in the USA. Recently, the family was expanded with several previously unknown arteriviruses isolated from different African monkey species. At the molecular level, arteriviruses share an intriguing but distant evolutionary relationship with coronaviruses and other members of the order Nidovirales. Nevertheless, several of their characteristics are unique, including virion composition and structure, and the conservation of only a subset of the replicase domains encountered in nidoviruses with larger genomes. During the past 15 years, the advent of reverse genetics systems for EAV and PRRSV has changed and accelerated the structure-function analysis of arterivirus RNA and protein sequences. These systems now also facilitate studies into host immune responses and arterivirus immune evasion and pathogenesis. In this review, we have summarized recent advances in the areas of arterivirus genome expression, RNA and protein functions, virion architecture, virus-host interactions, immunity, and pathogenesis. We have also briefly reviewed the impact of these advances on disease management, the engineering of novel candidate live vaccines and the diagnosis of arterivirus infection.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/genetics , Arterivirus/pathogenicity , Animals , Arterivirus/classification , Arterivirus/physiology , Arterivirus Infections/epidemiology , Arterivirus Infections/virology , Genome, Viral/genetics , Genome, Viral/physiology , Global Health , Mammals , Phylogeny
16.
Viruses ; 4(6): 980-1010, 2012 06.
Article in English | MEDLINE | ID: mdl-22816036

ABSTRACT

Animal coronaviruses, such as infectious bronchitis virus (IBV), and arteriviruses, such as porcine reproductive and respiratory syndrome virus (PRRSV), are able to manifest highly contagious infections in their specific native hosts, thereby arising in critical economic damage to animal industries. This review discusses recent progress in studies of virus-host interactions during animal and human coronavirus and arterivirus infections, with emphasis on IBV-host cell interactions. These interactions may be directly involved in viral replication or lead to the alteration of certain signaling pathways, such as cell stress response and innate immunity, to facilitate viral replication and pathogenesis.


Subject(s)
Arterivirus Infections/immunology , Arterivirus Infections/metabolism , Arterivirus/physiology , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Coronavirus/physiology , Host-Pathogen Interactions , Animals , Apoptosis , Cell Cycle , Humans , Immunity, Innate , Protein Biosynthesis , Signal Transduction
17.
J Virol ; 86(5): 2474-87, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190716

ABSTRACT

Virus-induced membrane structures support the assembly and function of positive-strand RNA virus replication complexes. The replicase proteins of arteriviruses are associated with double-membrane vesicles (DMVs), which were previously proposed to derive from the endoplasmic reticulum (ER). Using electron tomography, we performed an in-depth ultrastructural analysis of cells infected with the prototypic arterivirus equine arteritis virus (EAV). We established that the outer membranes of EAV-induced DMVs are interconnected with each other and with the ER, thus forming a reticulovesicular network (RVN) resembling that previously described for the distantly related severe acute respiratory syndrome (SARS) coronavirus. Despite significant morphological differences, a striking parallel between the two virus groups, and possibly all members of the order Nidovirales, is the accumulation in the DMV interior of double-stranded RNA, the presumed intermediate of viral RNA synthesis. In our electron tomograms, connections between the DMV interior and cytosol could not be unambiguously identified, suggesting that the double-stranded RNA is compartmentalized by the DMV membranes. As a novel approach to visualize and quantify the RNA content of viral replication structures, we explored electron spectroscopic imaging of DMVs, which revealed the presence of phosphorus in amounts equaling on average a few dozen copies of the EAV RNA genome. Finally, our electron tomograms revealed a network of nucleocapsid protein-containing protein tubules that appears to be intertwined with the RVN. This potential intermediate in nucleocapsid formation, which was not observed in coronavirus-infected cells, suggests that arterivirus RNA synthesis and assembly are coordinated in intracellular space.


Subject(s)
Arterivirus Infections/virology , Endoplasmic Reticulum/virology , Equartevirus/physiology , RNA, Viral/genetics , Virus Replication , Animals , Arterivirus/genetics , Arterivirus/physiology , Arterivirus/ultrastructure , Cell Line , Endoplasmic Reticulum/ultrastructure , Equartevirus/genetics , Equartevirus/ultrastructure , Intracellular Membranes/ultrastructure , Intracellular Membranes/virology , RNA, Viral/metabolism
18.
J Virol ; 82(9): 4480-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18305048

ABSTRACT

The replication/transcription complex of the arterivirus equine arteritis virus (EAV) is associated with paired membranes and/or double-membrane vesicles (DMVs) that are thought to originate from the endoplasmic reticulum. Previously, coexpression of two putative transmembrane nonstructural proteins (nsp2 and nsp3) was found to suffice to induce these remarkable membrane structures, which are typical of arterivirus infection. Here, site-directed mutagenesis was used to investigate the role of nsp3 in more detail. Liberation of the hydrophobic N terminus of nsp3, which is normally achieved by cleavage of the nsp2/3 junction by the nsp2 protease, was nonessential for the formation of DMVs. However, the substitution of each of a cluster of four conserved cysteine residues, residing in a predicted luminal loop of nsp3, completely blocked DMV formation. Some of these mutant nsp3 proteins were also found to be highly cytotoxic, in particular, exerting a dramatic effect on the endoplasmic reticulum. The functionality of an engineered N glycosylation site in the cysteine-containing loop confirmed both its presence in the lumen and the transmembrane nature of nsp3. This mutant displayed an interesting intermediate phenotype in terms of DMV formation, with paired and curved membranes being formed, but DMV formation apparently being impaired. The effect of nsp3 mutations on replicase polyprotein processing was investigated, and several mutations were found to influence processing of the region downstream of nsp3 by the nsp4 main protease. When tested in an EAV reverse genetics system, none of the nsp3 mutations was tolerated, again underlining the crucial role of the protein in the arterivirus life cycle.


Subject(s)
Arterivirus/chemistry , Intracellular Membranes/virology , Viral Nonstructural Proteins/physiology , Animals , Arterivirus/physiology , Arterivirus/ultrastructure , Horses , Multiprotein Complexes , Mutagenesis, Site-Directed , Transcription, Genetic , Viral Nonstructural Proteins/genetics , Virus Replication
19.
Virus Res ; 109(2): 109-24, 2005 May.
Article in English | MEDLINE | ID: mdl-15763141

ABSTRACT

Both full-length and subgenomic negative-strand RNAs are initiated at the 3' terminus of the positive-strand genomic RNA of the arterivirus, simian hemorrhagic fever virus (SHFV). The SHFV 3'(+) non-coding region (NCR) is 76 nts in length and forms a stem loop (SL) structure that was confirmed by ribonuclease structure probing. Two cell proteins, p56 and p42, bound specifically to a probe consisting of the SHFV 3'(+)NCR RNA. The 3'(+)NCR RNAs of two additional members of the arterivirus genus specifically interacted with two cell proteins of the same size. p56 was identified as polypyrimidine tract-binding protein (PTB) and p42 was identified as fructose bisphosphate aldolase A. PTB binding sites were mapped to a terminal loop and to a bulged region of the SHFV 3'SL structure. Deletion of either of the PTB binding sites in the viral RNA significantly reduced PTB binding activity, suggesting that both sites are required for efficient binding of this protein. Changes in the top portion of the SHFV 3'SL structure eliminated aldolase binding, suggesting that the binding site for this protein is located near the top of the SL. These cell proteins may play roles in regulating the functions of the genomic 3' NCR.


Subject(s)
Arterivirus/physiology , Fructose-Bisphosphate Aldolase/isolation & purification , Polypyrimidine Tract-Binding Protein/isolation & purification , RNA, Viral/metabolism , RNA-Binding Proteins/isolation & purification , Animals , Base Sequence , Cells, Cultured , Fructose-Bisphosphate Aldolase/metabolism , Macaca mulatta , Models, Molecular , Molecular Sequence Data , Molecular Weight , Nucleic Acid Conformation , Polypyrimidine Tract-Binding Protein/metabolism , Protein Binding , RNA, Untranslated/chemistry , RNA, Untranslated/metabolism , RNA, Viral/chemistry , RNA-Binding Proteins/metabolism , Sequence Deletion/genetics , Sequence Deletion/physiology
20.
Virology ; 303(2): 364-73, 2002 Nov 25.
Article in English | MEDLINE | ID: mdl-12490397

ABSTRACT

Arteriviruses are enveloped, positive-strand RNA viruses for which the two major envelope proteins GP(5) and M occur as disulfide-linked heterodimers. These were assumed to serve the viral targeting functions, but recent ectodomain swapping studies with equine arteritis virus (EAV) indicate that the GP(5) protein does not determine arteriviral tropism. Here, we focused on the short, 13- to 18-residue ectodomain of the M protein. Using an infectious cDNA clone of the Lelystad virus isolate of porcine reproductive and respiratory syndrome virus (PRRSV), we substituted the genomic sequence encoding the M ectodomain by that of murine lactate dehydrogenase-elevating virus, EAV, and the US PRRSV-isolate, VR2332. Viable viruses with a chimeric M protein were obtained in all three cases, but for the latter two only after removal of the genomic overlap between the M and GP(5) genes. Characterization of the chimeric viruses revealed that they could be distinguished immunologically from wild-type virus, that they were genetically stable in vitro, but that they were impaired in their growth, reaching lower titers than the parental virus. The latter appeared to be due to an increased particle-to-infectivity ratio of the chimeric virus particles. Interestingly, the chimeric viruses had retained their ability to infect porcine cells and had not acquired tropism for cells susceptible to the viruses from which the foreign ectodomains were derived. We conclude that the surface structures composed by the arterivirus M and GP(5) ectodomains do not determine viral tropism.


Subject(s)
Arterivirus/physiology , Recombinant Fusion Proteins/physiology , Viral Matrix Proteins/physiology , Amino Acid Sequence , Animals , Arterivirus/genetics , Arterivirus/immunology , Base Sequence , Equartevirus/physiology , Lactate dehydrogenase-elevating virus/physiology , Molecular Sequence Data , Open Reading Frames , Porcine respiratory and reproductive syndrome virus/physiology , Swine , Transfection , Viral Envelope Proteins/physiology , Viral Matrix Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...