Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Virol ; 90(9): 4357-4368, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26889024

RESUMO

UNLABELLED: The coronavirus membrane (M) protein is the central actor in virion morphogenesis. M organizes the components of the viral membrane, and interactions of M with itself and with the nucleocapsid (N) protein drive virus assembly and budding. In order to further define M-M and M-N interactions, we constructed mutants of the model coronavirus mouse hepatitis virus (MHV) in which all or part of the M protein was replaced by its phylogenetically divergent counterpart from severe acute respiratory syndrome coronavirus (SARS-CoV). We were able to obtain viable chimeras containing the entire SARS-CoV M protein as well as mutants with intramolecular substitutions that partitioned M protein at the boundaries between the ectodomain, transmembrane domains, or endodomain. Our results show that the carboxy-terminal domain of N protein, N3, is necessary and sufficient for interaction with M protein. However, despite some previous genetic and biochemical evidence that mapped interactions with N to the carboxy terminus of M, it was not possible to define a short linear region of M protein sufficient for assembly with N. Thus, interactions with N protein likely involve multiple linearly discontiguous regions of the M endodomain. The SARS-CoV M chimera exhibited a conditional growth defect that was partially suppressed by mutations in the envelope (E) protein. Moreover, virions of the M chimera were markedly deficient in spike (S) protein incorporation. These findings suggest that the interactions of M protein with both E and S protein are more complex than previously thought. IMPORTANCE: The assembly of coronavirus virions entails concerted interactions among the viral structural proteins and the RNA genome. One strategy to study this process is through construction of interspecies chimeras that preserve or disrupt particular inter- or intramolecular associations. In this work, we replaced the membrane (M) protein of the model coronavirus mouse hepatitis virus with its counterpart from a heterologous coronavirus. The results clarify our understanding of the interaction between the coronavirus M protein and the nucleocapsid protein. At the same time, they reveal unanticipated complexities in the interactions of M with the viral spike and envelope proteins.


Assuntos
Infecções por Coronavirus/virologia , Coronavirus/fisiologia , Proteínas do Nucleocapsídeo/metabolismo , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus , Sequência de Aminoácidos , Animais , Linhagem Celular , Proteínas M de Coronavírus , Proteínas do Nucleocapsídeo de Coronavírus , Vetores Genéticos/genética , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/fisiologia , Mutação , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética , Vírion , Replicação Viral
2.
J Virol ; 89(11): 6033-47, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25810552

RESUMO

UNLABELLED: Coronaviruses, the largest RNA viruses, have a complex program of RNA synthesis that entails genome replication and transcription of subgenomic mRNAs. RNA synthesis by the prototype coronavirus mouse hepatitis virus (MHV) is carried out by a replicase-transcriptase composed of 16 nonstructural protein (nsp) subunits. Among these, nsp3 is the largest and the first to be inserted into the endoplasmic reticulum. nsp3 comprises multiple structural domains, including two papain-like proteases (PLPs) and a highly conserved ADP-ribose-1″-phosphatase (ADRP) macrodomain. We have previously shown that the ubiquitin-like domain at the amino terminus of nsp3 is essential and participates in a critical interaction with the viral nucleocapsid protein early in infection. In the current study, we exploited atypical expression schemes to uncouple PLP1 from the processing of nsp1 and nsp2 in order to investigate the requirements of nsp3 domains for viral RNA synthesis. In the first strategy, a mutant was created in which replicase polyprotein translation initiated with nsp3, thereby establishing that complete elimination of nsp1 and nsp2 does not abolish MHV viability. In the second strategy, a picornavirus autoprocessing element was used to separate a truncated nsp1 from nsp3. This provided a platform for further dissection of amino-terminal domains of nsp3. From this, we found that catalytic mutation of PLP1 or complete deletion of PLP1 and the adjacent ADRP domain was tolerated by the virus. These results showed that neither the PLP1 domain nor the ADRP domain of nsp3 provides integral activities essential for coronavirus genomic or subgenomic RNA synthesis. IMPORTANCE: The largest component of the coronavirus replicase-transcriptase complex, nsp3, contains multiple modules, many of which do not have clearly defined functions in genome replication or transcription. These domains may play direct roles in RNA synthesis, or they may have evolved for other purposes, such as to combat host innate immunity. We initiated a dissection of MHV nsp3 aimed at identifying those activities or structures in this huge molecule that are essential to replicase activity. We found that both PLP1 and ADRP could be entirely deleted, provided that the requirement for proteolytic processing by PLP1 was offset by an alternative mechanism. This demonstrated that neither PLP1 nor ADRP plays an essential role in coronavirus RNA synthesis.


Assuntos
Vírus da Hepatite Murina/fisiologia , RNA Viral/biossíntese , Transcrição Gênica , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Análise Mutacional de DNA , Camundongos , Viabilidade Microbiana , Vírus da Hepatite Murina/genética , Estrutura Terciária de Proteína , Proteínas não Estruturais Virais/genética
3.
J Virol ; 88(8): 4451-65, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24501403

RESUMO

UNLABELLED: The coronavirus nucleocapsid (N) protein forms a helical ribonucleoprotein with the viral positive-strand RNA genome and binds to the principal constituent of the virion envelope, the membrane (M) protein, to facilitate assembly and budding. Besides these structural roles, N protein associates with a component of the replicase-transcriptase complex, nonstructural protein 3, at a critical early stage of infection. N protein has also been proposed to participate in the replication and selective packaging of genomic RNA and the transcription and translation of subgenomic mRNA. Coronavirus N proteins contain two structurally distinct RNA-binding domains, an unusual characteristic among RNA viruses. To probe the functions of these domains in the N protein of the model coronavirus mouse hepatitis virus (MHV), we constructed mutants in which each RNA-binding domain was replaced by its counterpart from the N protein of severe acute respiratory syndrome coronavirus (SARS-CoV). Mapping of revertants of the resulting chimeric viruses provided evidence for extensive intramolecular interactions between the two RNA-binding domains. Through analysis of viral RNA that was packaged into virions we identified the second of the two RNA-binding domains as a principal determinant of MHV packaging signal recognition. As expected, the interaction of N protein with M protein was not affected in either of the chimeric viruses. Moreover, the SARS-CoV N substitutions did not alter the fidelity of leader-body junction formation during subgenomic mRNA synthesis. These results more clearly delineate the functions of N protein and establish a basis for further exploration of the mechanism of genomic RNA packaging. IMPORTANCE: This work describes the interactions of the two RNA-binding domains of the nucleocapsid protein of a model coronavirus, mouse hepatitis virus. The main finding is that the second of the two domains plays an essential role in recognizing the RNA structure that allows the selective packaging of genomic RNA into assembled virions.


Assuntos
Infecções por Coronaviridae/veterinária , Genoma Viral , Vírus da Hepatite Murina/fisiologia , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/metabolismo , RNA Viral/metabolismo , Doenças dos Roedores/virologia , Montagem de Vírus , Animais , Linhagem Celular , Infecções por Coronaviridae/virologia , Proteínas do Nucleocapsídeo de Coronavírus , Camundongos , Vírus da Hepatite Murina/química , Vírus da Hepatite Murina/genética , Proteínas do Nucleocapsídeo/genética , Ligação Proteica , Estrutura Terciária de Proteína , RNA Viral/genética
4.
J Virol ; 87(9): 5182-92, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23449786

RESUMO

Coronaviruses selectively package genomic RNA into assembled virions, despite the great molar excess of subgenomic RNA species that is present in infected cells. The genomic packaging signal (PS) for the coronavirus mouse hepatitis virus (MHV) was originally identified as an element that conferred packaging capability to defective interfering RNAs. The MHV PS is an RNA structure that maps to the region of the replicase gene encoding the nonstructural protein 15 subunit of the viral replicase-transcriptase complex. To begin to understand the role and mechanism of action of the MHV PS in its native genomic locus, we constructed viral mutants in which this cis-acting element was altered, deleted, or transposed. Our results demonstrated that the PS is pivotal in the selection of viral genomic RNA for incorporation into virions. Mutants in which PS RNA secondary structure was disrupted or entirely ablated packaged large quantities of subgenomic RNAs, in addition to genomic RNA. Moreover, the PS retained its function when displaced to an ectopic site in the genome. Surprisingly, the PS was not essential for MHV viability, nor did its elimination have a severe effect on viral growth. However, the PS was found to provide a distinct selective advantage to MHV. Viruses containing the PS readily outcompeted their otherwise isogenic counterparts lacking the PS.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/genética , Genoma Viral , RNA Viral/química , RNA Viral/genética , Doenças dos Roedores/virologia , Montagem de Vírus , Sequência de Aminoácidos , Animais , Sequência de Bases , Coronaviridae/química , Coronaviridae/fisiologia , Infecções por Coronaviridae/virologia , Camundongos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Viral/metabolismo , Vírion/química , Vírion/genética , Vírion/fisiologia
5.
J Virol ; 87(16): 9159-72, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23760243

RESUMO

The coronavirus nucleocapsid protein (N) plays an essential structural role in virions through a network of interactions with positive-strand viral genomic RNA, the envelope membrane protein (M), and other N molecules. Additionally, N protein participates in at least one stage of the complex mechanism of coronavirus RNA synthesis. We previously uncovered an unanticipated interaction between N and the largest subunit of the viral replicase-transcriptase complex, nonstructural protein 3 (nsp3). This was found through analysis of revertants of a severely defective mutant of murine hepatitis virus (MHV) in which the N gene was replaced with that of its close relative, bovine coronavirus (BCoV). In the work reported here, we constructed BCoV chimeras and other mutants of MHV nsp3 and obtained complementary genetic evidence for its association with N protein. We found that the N-nsp3 interaction maps to the amino-terminal ubiquitin-like domain of nsp3, which is essential for the virus. The interaction does not require the adjacent acidic domain of nsp3, which is dispensable. In addition, we demonstrated a complete correspondence between N-nsp3 genetic interactions and the ability of N protein to enhance the infectivity of transfected coronavirus genomic RNA. The latter function of N was shown to depend on both of the RNA-binding domains of N, as well as on the serine- and arginine-rich central region of N, which binds nsp3. Our results support a model in which the N-nsp3 interaction serves to tether the genome to the newly translated replicase-transcriptase complex at a very early stage of infection.


Assuntos
Coronavirus Bovino/fisiologia , Vírus da Hepatite Murina/fisiologia , Proteínas do Nucleocapsídeo/metabolismo , Mapeamento de Interação de Proteínas , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Coronavirus Bovino/genética , Camundongos , Vírus da Hepatite Murina/genética , Proteínas do Nucleocapsídeo/genética , Recombinação Genética , Transgenes , Proteínas não Estruturais Virais/genética
6.
Virology ; 567: 1-14, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34933176

RESUMO

The coronavirus nucleocapsid (N) protein comprises two RNA-binding domains connected by a central spacer, which contains a serine- and arginine-rich (SR) region. The SR region engages the largest subunit of the viral replicase-transcriptase, nonstructural protein 3 (nsp3), in an interaction that is essential for efficient initiation of infection by genomic RNA. We carried out an extensive genetic analysis of the SR region of the N protein of mouse hepatitis virus in order to more precisely define its role in RNA synthesis. We further examined the N-nsp3 interaction through construction of nsp3 mutants and by creation of an interspecies N protein chimera. Our results indicate a role for the central spacer as an interaction hub of the N molecule that is partially regulated by phosphorylation. These findings are discussed in relation to the recent discovery that nsp3 forms a molecular pore in the double-membrane vesicles that sequester the coronavirus replicase-transcriptase.


Assuntos
Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Membranas Intracelulares/metabolismo , Proteínas do Complexo da Replicase Viral/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular , Proteínas do Nucleocapsídeo de Coronavírus/química , Proteínas do Nucleocapsídeo de Coronavírus/genética , RNA-Polimerase RNA-Dependente de Coronavírus/química , RNA-Polimerase RNA-Dependente de Coronavírus/genética , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , Camundongos , Vírus da Hepatite Murina , Mutação , Ligação Proteica , Domínios Proteicos , RNA Viral/biossíntese , Proteínas do Complexo da Replicase Viral/química , Proteínas do Complexo da Replicase Viral/genética , Compartimentos de Replicação Viral/metabolismo
7.
J Virol ; 84(24): 12872-85, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926558

RESUMO

The coronavirus small envelope (E) protein plays a crucial, but poorly defined, role in the assembly of virions. To investigate E protein function, we previously generated E gene point mutants of mouse hepatitis virus (MHV) that were defective in growth and assembled virions with anomalous morphologies. We subsequently constructed an E gene deletion (ΔE) mutant that was only minimally viable. The ΔE virus formed tiny plaques and reached optimal infectious titers many orders of magnitude below those of wild-type virus. We have now characterized highly aberrant viral transcription patterns that developed in some stocks of the ΔE mutant. Extensive analysis of three independent stocks revealed that, in each, a faster-growing virus harboring a genomic duplication had been selected. Remarkably, the net result of each duplication was the creation of a variant version of the membrane protein (M) gene that was situated upstream of the native copy of the M gene. Each different variant M gene encoded an expressed protein (M*) containing a truncated endodomain. Reconstruction of one variant M gene in a ΔE background showed that expression of the M* protein markedly enhanced the growth of the ΔE mutant and that the M* protein was incorporated into assembled virions. These findings suggest that M* proteins were repeatedly selected as surrogates for the E protein and that one role of E is to mediate interactions between transmembrane domains of M protein monomers. Our results provide a demonstration of the capability of coronaviruses to evolve new gene functions through recombination.


Assuntos
Evolução Biológica , Proteínas de Membrana/genética , Vírus da Hepatite Murina/genética , Proteínas do Envelope Viral/genética , Vírion/fisiologia , Montagem de Vírus/fisiologia , Sequência de Aminoácidos , Animais , Northern Blotting , Western Blotting , Linhagem Celular , DNA Viral/genética , Genoma Viral , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/metabolismo , Mutação/genética , Fenótipo , RNA Viral/genética , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Proteínas do Envelope Viral/metabolismo
8.
J Virol ; 84(19): 10276-88, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660183

RESUMO

The coronavirus nucleocapsid (N) protein plays an essential role in virion assembly via interactions with the large, positive-strand RNA viral genome and the carboxy-terminal endodomain of the membrane protein (M). To learn about the functions of N protein domains in the coronavirus mouse hepatitis virus (MHV), we replaced the MHV N gene with its counterpart from the closely related bovine coronavirus (BCoV). The resulting viral mutant was severely defective, even though individual domains of the N protein responsible for N-RNA, N-M, or N-N interactions were completely interchangeable between BCoV and MHV. The lesion in the BCoV N substitution mutant could be compensated for by reverting mutations in the central, serine- and arginine-rich (SR) domain of the N protein. Surprisingly, a second class of reverting mutations were mapped to the amino terminus of a replicase subunit, nonstructural protein 3 (nsp3). A similarly defective MHV N mutant bearing an insertion of the SR region from the severe acute respiratory syndrome coronavirus N protein was rescued by the same two classes of reverting mutations. Our genetic results were corroborated by the demonstration that the expressed amino-terminal segment of nsp3 bound selectively to N protein from infected cells, and this interaction was RNA independent. Moreover, we found a direct correlation between the N-nsp3 interaction and the ability of N protein to stimulate the infectivity of transfected MHV genomic RNA (gRNA). Our results suggest a role for this previously unknown N-nsp3 interaction in the localization of genomic RNA to the replicase complex at an early stage of infection.


Assuntos
Coronavirus Bovino/fisiologia , RNA Polimerases Dirigidas por DNA/fisiologia , Vírus da Hepatite Murina/fisiologia , Proteínas do Nucleocapsídeo/fisiologia , RNA Viral/genética , RNA Polimerase Dependente de RNA/fisiologia , Sequência de Aminoácidos , Animais , Bovinos , Proteínas do Nucleocapsídeo de Coronavírus , Coronavirus Bovino/genética , Coronavirus Bovino/patogenicidade , RNA Polimerases Dirigidas por DNA/genética , Genoma Viral , Humanos , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/patogenicidade , Mutação , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/genética , Estrutura Terciária de Proteína , RNA Polimerase Dependente de RNA/genética , Recombinação Genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Homologia de Sequência de Aminoácidos , Transfecção , Virulência/genética , Virulência/fisiologia
9.
J Virol ; 84(16): 8262-74, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519394

RESUMO

The type I interferon (IFN) response plays an essential role in the control of in vivo infection by the coronavirus mouse hepatitis virus (MHV). However, in vitro, most strains of MHV are largely resistant to the action of this cytokine, suggesting that MHV encodes one or more functions that antagonize or evade the IFN system. A particular strain of MHV, MHV-S, exhibited orders-of-magnitude higher sensitivity to IFN than prototype strain MHV-A59. Through construction of interstrain chimeric recombinants, the basis for the enhanced IFN sensitivity of MHV-S was found to map entirely to the region downstream of the spike gene, at the 3' end of the genome. Sequence analysis revealed that the major difference between the two strains in this region is the absence of gene 5a from MHV-S. Creation of a gene 5a knockout mutant of MHV-A59 demonstrated that a major component of IFN resistance maps to gene 5a. Conversely, insertion of gene 5a, or its homologs from related group 2 coronaviruses, at an upstream genomic position in an MHV-A59/S chimera restored IFN resistance. This is the first demonstration of a coronavirus gene product that can protect that same virus from the antiviral state induced by IFN. Neither protein kinase R, which phosphorylates eukaryotic initiation factor 2, nor oligoadenylate synthetase, which activates RNase L, was differentially activated in IFN-treated cells infected with MHV-A59 or MHV-S. Thus, the major IFN-induced antiviral activities that are specifically inhibited by MHV, and possibly by other coronaviruses, remain to be identified.


Assuntos
Interferons/antagonistas & inibidores , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/patogenicidade , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Mapeamento Cromossômico , Análise Mutacional de DNA , Técnicas de Inativação de Genes , Teste de Complementação Genética , Evasão da Resposta Imune , Tolerância Imunológica , Interferons/imunologia , Camundongos , Dados de Sequência Molecular , RNA Viral/genética , Análise de Sequência de DNA , Ensaio de Placa Viral , Proteínas Virais/imunologia , Fatores de Virulência/imunologia
10.
J Virol ; 84(21): 11575-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20739524

RESUMO

The coronavirus nucleocapsid (N) protein is a virion structural protein. It also functions, however, in an unknown way in viral replication and localizes to the viral replication-transcription complexes (RTCs). Here we investigated, using recombinant murine coronaviruses expressing green fluorescent protein (GFP)-tagged versions of the N protein, the dynamics of its interactions with the RTCs and the domain(s) involved. Using fluorescent recovery after photobleaching, we showed that the N protein, unlike the nonstructural protein 2, is dynamically associated with the RTCs. Recruitment of the N protein to the RTCs requires the C-terminal N2b domain, which interacts with other N proteins in an RNA-independent manner.


Assuntos
Proteínas do Nucleocapsídeo/metabolismo , Transcrição Gênica , Replicação Viral , Animais , Proteínas do Nucleocapsídeo de Coronavírus , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico
11.
J Virol ; 83(14): 7221-34, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19420077

RESUMO

The coronavirus nucleocapsid protein (N), together with the large, positive-strand RNA viral genome, forms a helically symmetric nucleocapsid. This ribonucleoprotein structure becomes packaged into virions through association with the carboxy-terminal endodomain of the membrane protein (M), which is the principal constituent of the virion envelope. Previous work with the prototype coronavirus mouse hepatitis virus (MHV) has shown that a major determinant of the N-M interaction maps to the carboxy-terminal domain 3 of the N protein. To explore other domain interactions of the MHV N protein, we expressed a series of segments of the MHV N protein as fusions with green fluorescent protein (GFP) during the course of viral infection. We found that two of these GFP-N-domain fusion proteins were selectively packaged into virions as the result of tight binding to the N protein in the viral nucleocapsid, in a manner that did not involve association with either M protein or RNA. The nature of each type of binding was further explored through genetic analysis. Our results defined two strongly interacting regions of the N protein. One is the same domain 3 that is critical for M protein recognition during assembly. The other is domain N1b, which corresponds to the N-terminal domain that has been structurally characterized in detail for two other coronaviruses, infectious bronchitis virus and the severe acute respiratory syndrome coronavirus.


Assuntos
Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/veterinária , Vírus da Hepatite Murina/fisiologia , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/metabolismo , Doenças dos Roedores/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/virologia , Infecções por Coronavirus/virologia , Proteínas do Nucleocapsídeo de Coronavírus , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/química , Vírus da Hepatite Murina/genética , Mutação , Proteínas do Nucleocapsídeo/genética , Ligação Proteica , Estrutura Terciária de Proteína , Doenças dos Roedores/virologia , Montagem de Vírus
12.
Sci Rep ; 10(1): 13893, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32807800

RESUMO

The psychrophilic (cold-loving) fungus Pseudogymnoascus destructans was discovered more than a decade ago to be the pathogen responsible for white-nose syndrome, an emerging disease of North American bats causing unprecedented population declines. The same species of fungus is found in Europe but without associated mortality in bats. We found P. destructans was infected with a mycovirus [named Pseudogymnoascus destructans partitivirus 1 (PdPV-1)]. The virus is bipartite, containing two double-stranded RNA (dsRNA) segments designated as dsRNA1 and dsRNA2. The cDNA sequences revealed that dsRNA1 dsRNA is 1,683 bp in length with an open reading frame (ORF) that encodes 539 amino acids (molecular mass of 62.7 kDa); dsRNA2 dsRNA is 1,524 bp in length with an ORF that encodes 434 amino acids (molecular mass of 46.9 kDa). The dsRNA1 ORF contains motifs representative of RNA-dependent RNA polymerase (RdRp), whereas the dsRNA2 ORF sequence showed homology with the putative capsid proteins (CPs) of mycoviruses. Phylogenetic analyses with PdPV-1 RdRp and CP sequences indicated that both segments constitute the genome of a novel virus in the family Partitiviridae. The purified virions were isometric with an estimated diameter of 33 nm. Reverse transcription PCR (RT-PCR) and sequencing revealed that all US isolates and a subset of Czech Republic isolates of P. destructans were infected with PdPV-1. However, PdPV-1 appears to be not widely dispersed in the fungal genus Pseudogymnoascus, as non-pathogenic fungi P. appendiculatus (1 isolate) and P. roseus (6 isolates) tested negative. P. destructans PdPV-1 could be a valuable tool to investigate fungal biogeography and the host-pathogen interactions in bat WNS.


Assuntos
Ascomicetos/virologia , Quirópteros/microbiologia , Micovírus/fisiologia , Sequência de Aminoácidos , Animais , Ascomicetos/isolamento & purificação , Sequência de Bases , República Tcheca , Micovírus/genética , Micovírus/ultraestrutura , Filogenia , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/isolamento & purificação , RNA Viral/genética , RNA Viral/isolamento & purificação , Estados Unidos , Proteínas Virais/química
13.
J Virol ; 82(3): 1214-28, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18032506

RESUMO

The upstream end of the 3' untranslated region (UTR) of the mouse hepatitis virus genome contains two essential and overlapping RNA secondary structures, a bulged stem-loop and a pseudoknot, which have been proposed to be elements of a molecular switch that is critical for viral RNA synthesis. It has previously been shown that a particular six-base insertion in loop 1 of the pseudoknot is extremely deleterious to the virus. We have now isolated multiple independent second-site revertants of the loop 1 insertion mutant, and we used reverse-genetics methods to confirm the identities of suppressor mutations that could compensate for the original insertion. The suppressors were localized to two separate regions of the genome. Members of one class of suppressor were mapped to the portions of gene 1 that encode nsp8 and nsp9, thereby providing the first evidence for specific interactions between coronavirus replicase gene products and a cis-acting genomic RNA element. The second class of suppressor was mapped to the extreme 3' end of the genome, a result which pointed to the existence of a direct base-pairing interaction between loop 1 of the pseudoknot and the genomic terminus. The latter finding was strongly supported by phylogenetic evidence and by the construction of a deletion mutant that reduced the 3' UTR to its minimal essential elements. Taken together, the interactions revealed by the two classes of suppressors suggest a model for the initiation of coronavirus negative-strand RNA synthesis.


Assuntos
Regiões 3' não Traduzidas/metabolismo , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/metabolismo , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Regiões 3' não Traduzidas/química , Regiões 3' não Traduzidas/genética , Animais , Camundongos , Conformação de Ácido Nucleico , RNA Viral/química , RNA Viral/genética , RNA Polimerase Dependente de RNA/genética , Supressão Genética , Transcrição Gênica , Replicação Viral/fisiologia
14.
Virology ; 537: 198-207, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31505321

RESUMO

RNA viruses carry out selective packaging of their genomes in a variety of ways, many involving a genomic packaging signal. The first coronavirus packaging signal was discovered nearly thirty years ago, but how it functions remains incompletely understood. This review addresses the current state of knowledge of coronavirus genome packaging, which has mainly been studied in two prototype species, mouse hepatitis virus and transmissible gastroenteritis virus. Despite the progress that has been made in the mapping and characterization of some packaging signals, there is conflicting evidence as to whether the viral nucleocapsid protein or the membrane protein plays the primary role in packaging signal recognition. The different models for the mechanism of genomic RNA packaging that have been prompted by these competing views are described. Also discussed is the recent exciting discovery that selective coronavirus genome packaging is critical for in vivo evasion of the host innate immune response.


Assuntos
Vírus da Hepatite Murina/fisiologia , RNA Viral/metabolismo , Vírus da Gastroenterite Transmissível/fisiologia , Montagem de Vírus , Modelos Biológicos , Proteínas do Nucleocapsídeo/metabolismo , Proteínas da Matriz Viral/metabolismo
15.
Methods Mol Biol ; 454: 229-36, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19057874

RESUMO

Targeted RNA recombination has proven to be a powerful tool for the genetic engineering of the coronavirus genome, particularly in its 3' part. Here we describe procedures for the generation of recombinant and mutant mouse hepatitis virus and feline infectious peritonitis virus. Key to the two-step method is the efficient selection of recombinant viruses based on host cell switching. The first step consists of the preparation---using this selection principle--of an interspecies chimeric coronavirus. In this virus the ectodomain of the spike glycoprotein is replaced by that of a coronavirus with a different species tropism. In the second step this chimeric virus is used as the recipient for recombination with synthetic donor RNA carrying the original spike gene. Recombinant viruses are then isolated on the basis of their regained natural (e.g., murine or feline) cell tropism. Additional mutations created in the donor RNA can be co-incorporated into the recombinant virus in order to generate mutant viruses.


Assuntos
Coronavirus/genética , Genoma Viral/genética , RNA Viral/genética , Recombinação Genética , Animais , Gatos , Coronavirus Felino/genética , Camundongos , Modelos Genéticos , Vírus da Hepatite Murina/genética
16.
Virology ; 494: 100-7, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27105451

RESUMO

The prototype coronavirus mouse hepatitis virus (MHV) exhibits highly selective packaging of its genomic positive-stranded RNA into assembled virions, despite the presence in infected cells of a large excess of subgenomic viral mRNAs. One component of this selectivity is the MHV packaging signal (PS), an RNA structure found only in genomic RNA and not in subgenomic RNAs. It was previously shown that a major determinant of PS recognition is the second of the two RNA-binding domains of the viral nucleocapsid (N) protein. We have now found that PS recognition additionally depends upon a segment of the carboxy-terminal tail (domain N3) of the N protein. Since domain N3 is also the region of N protein that interacts with the membrane (M) protein, this finding suggests a mechanism by which selective genome packaging is accomplished, through the coupling of genome encapsidation to virion assembly.


Assuntos
Coronavirus/fisiologia , Genoma Viral , Proteínas do Nucleocapsídeo/metabolismo , Domínios e Motivos de Interação entre Proteínas , RNA Viral , Montagem de Vírus , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas do Nucleocapsídeo de Coronavírus , Ordem dos Genes , Camundongos , Modelos Biológicos , Conformação de Ácido Nucleico , Proteínas do Nucleocapsídeo/química , Ligação Proteica , Proteínas Recombinantes de Fusão
17.
Virology ; 442(1): 74-81, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-23628137

RESUMO

Coronavirus spike (S) protein assembles into virions via its carboxy-terminus, which is composed of a transmembrane domain and an endodomain. Here, the carboxy-terminal charge-rich motif in the endodomain was verified to be critical for the specificity of S assembly into mouse hepatitis virus (MHV). Recombinant MHVs exhibited a range of abilities to accommodate the homologous S endodomains from the betacoronaviruses bovine coronavirus and human SARS-associated coronavirus, the alphacoronavirus porcine transmissible gastroenteritis virus (TGEV), and the gammacoronavirus avian infectious bronchitis virus respectively. Interestingly, in TGEV endodomain chimeras the reverting mutations resulted in stronger S incorporation into virions, and a net gain of negatively charged residues in the charge-rich motif accounted for the improvement. Additionally, MHV S assembly could also be rescued by the acidic carboxy-terminal domain of the nucleocapsid protein. These results indicate an important role for negatively charged endodomain residues in the incorporation of MHV S protein into assembled virions.


Assuntos
Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Vírus da Hepatite Murina/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Montagem de Vírus , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Coronavirus Bovino/genética , Coronavirus Bovino/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/química , Vírus da Hepatite Murina/genética , Mutação , Estrutura Terciária de Proteína/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Glicoproteína da Espícula de Coronavírus , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/metabolismo , Proteínas do Envelope Viral/genética
19.
J Virol ; 81(5): 2249-62, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17182690

RESUMO

The small envelope protein (E) plays a role of central importance in the assembly of coronaviruses. This was initially established by studies demonstrating that cellular expression of only E protein and the membrane protein (M) was necessary and sufficient for the generation and release of virus-like particles. To investigate the role of E protein in the whole virus, we previously generated E gene mutants of mouse hepatitis virus (MHV) that were defective in viral growth and produced aberrantly assembled virions. Surprisingly, however, we were also able to isolate a viable MHV mutant (DeltaE) in which the entire E gene, as well as the nonessential upstream genes 4 and 5a, were deleted. We have now constructed an E knockout mutant that confirms that the highly defective phenotype of the DeltaE mutant is due to loss of the E gene. Additionally, we have created substitution mutants in which the MHV E gene was replaced by heterologous E genes from viruses spanning all three groups of the coronavirus family. Group 2 and 3 E proteins were readily exchangeable for that of MHV. However, the E protein of a group 1 coronavirus, transmissible gastroenteritis virus, became functional in MHV only after acquisition of particular mutations. Our results show that proteins encompassing a remarkably diverse range of primary amino acid sequences can provide E protein function in MHV. These findings suggest that E protein facilitates viral assembly in a manner that does not require E protein to make sequence-specific contacts with M protein.


Assuntos
Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Gatos , Linhagem Celular , DNA Viral/genética , Expressão Gênica , Genes Virais , Camundongos , Dados de Sequência Molecular , Mutação , Fenótipo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/fisiologia , Proteínas do Envelope Viral/química , Replicação Viral
20.
J Virol ; 81(3): 1274-87, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17093194

RESUMO

The 3' cis-acting element for mouse hepatitis virus (MHV) RNA synthesis resides entirely within the 301-nucleotide 3' untranslated region (3' UTR) of the viral genome and consists of three regions. Encompassing the upstream end of the 3' UTR are a bulged stem-loop and an overlapping RNA pseudoknot, both of which are essential to MHV and common to all group 2 coronaviruses. At the downstream end of the genome is the minimal signal for initiation of negative-strand RNA synthesis. Between these two ends is a hypervariable region (HVR) that is only poorly conserved between MHV and other group 2 coronaviruses. Paradoxically, buried within the HVR is an octanucleotide motif (oct), 5'-GGAAGAGC-3', which is almost universally conserved in coronaviruses and is therefore assumed to have a critical biological function. We conducted an extensive mutational analysis of the HVR. Surprisingly, this region tolerated numerous deletions, rearrangements, and point mutations. Most striking, a mutant deleted of the entire HVR was only minimally impaired in tissue culture relative to the wild type. By contrast, the HVR deletion mutant was highly attenuated in mice, causing no signs of clinical disease and minimal weight loss compared to wild-type virus. Correspondingly, replication of the HVR deletion mutant in the brains of mice was greatly reduced compared to that of the wild type. Our results show that neither the HVR nor oct is essential for the basic mechanism of MHV RNA synthesis in tissue culture. However, the HVR appears to play a significant role in viral pathogenesis.


Assuntos
Regiões 3' não Traduzidas/fisiologia , Vírus da Hepatite Murina/patogenicidade , RNA Viral/genética , Regiões 3' não Traduzidas/genética , Animais , Linhagem Celular , Camundongos , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/fisiologia , Conformação de Ácido Nucleico , RNA Viral/química , RNA Viral/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA