Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 75(15): 6800-7, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11435558

RESUMO

The Sendai virus (SeV) C gene codes for a nested set of four C proteins that carry out several functions, including the modulation of viral RNA synthesis and countering of the cellular antiviral response. Using mutant C genes (and in particular a C gene with a deletion of six amino acids present only in the larger pair of C proteins) and recombinant SeV carrying these mutant C genes, we find that the nested set of C proteins carry out a nested set of functions. All of the C proteins interdict interferon (IFN) signaling to IFN-stimulated genes (ISGs) and prevent pY701-Stat1 formation. However, only the larger C proteins can induce STAT1 instability, prevent IFN from inducing an antiviral state, or prevent programmed cell death. Remarkably, interdiction of IFN signaling to ISGs and the absence of pY701-Stat1 formation did not prevent IFN-alpha from inducing an anti-Vesicular stomatitis virus (VSV) state. It is possible that IFN-alpha signaling to induce an anti-VSV state can occur independently of the well-established Jak/Stat/ISGF3 pathway and that it is this parallel pathway that is targeted by the longer C proteins.


Assuntos
Respirovirus/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Linhagem Celular , Cricetinae , Efeito Citopatogênico Viral , Proteínas de Ligação a DNA/metabolismo , Genes Virais , Células HeLa , Humanos , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Janus Quinase 1 , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Proteínas Tirosina Quinases/metabolismo , RNA Viral/biossíntese , Respirovirus/metabolismo , Fator de Transcrição STAT1 , Transdução de Sinais , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
2.
J Virol ; 74(19): 8823-30, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10982324

RESUMO

Sendai virus (SeV) infection of interferon (IFN)-competent cells is one of the most efficient ways of inducing IFN production. Virus replication is nevertheless largely unaffected, since SeV infection also interfers with IFN action, a prerequisite for the establishment of an antiviral state. This property has been mapped by reverse genetics to the viral C gene, which is also known to act as a promoter-specific inhibitor of viral RNA synthesis. Using luciferase reporter plasmids containing IFN-responsive promoters, we have found that all four C proteins effectively interdict IFN signaling when expressed independently of SeV infection. The C proteins must therefore interact directly with cellular components to carry this out. The C gene in the context of an SeV infection was also found to induce STAT1 instability in some cells, whereas in other cells it apparently acts to prevent the synthesis of STAT1 in response to the virus infection or IFN treatment. The SeV C proteins appear to act in at least two ways to counteract the IFN induced by SeV infection.


Assuntos
Interferons/metabolismo , Infecções por Respirovirus/metabolismo , Respirovirus/fisiologia , Proteínas Virais/fisiologia , Animais , Interferons/farmacologia , Camundongos , Infecções por Respirovirus/virologia , Transdução de Sinais , Replicação Viral
3.
Adv Virus Res ; 54: 403-22, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10547681

RESUMO

Molecular studies on the replication of paramyxoviruses have undergone a revolution in recent years due to the development of techniques that permit the manipulation of their genomes as cDNA. This has led to new information on the structure-function organization of the viral proteins involved in genome expression, as well as dissection of the cis-acting template sequences that regulate transcription and replication. Studies using recombinant viruses have also provided new insights into the role of the accessory proteins (V, C, M1/M2) in both for virus growth in cultured cells and pathogenesis in animals.


Assuntos
Paramyxoviridae/genética , Paramyxoviridae/fisiologia , Replicação Viral , Transcrição Gênica , Proteínas Virais/genética , Proteínas Virais/metabolismo
4.
J Virol ; 73(8): 6559-65, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10400752

RESUMO

We have studied the relationship between the Sendai virus (SeV) C proteins (a nested set of four proteins initiated at different start codons) and the interferon (IFN)-mediated antiviral response in IFN-competent cells in culture. SeV strains containing wild-type or various mutant C proteins were examined for their ability (i) to induce an antiviral state (i.e., to prevent the growth of vesicular stomatitis virus [VSV] following a period of SeV infection), (ii) to induce the elevation of Stat1 protein levels, and (iii) to prevent IFN added concomitant with the SeV infection from inducing an antiviral state. We find that expression of the wild-type C gene and, specifically, the AUG114-initiated C protein prevents the establishment of an antiviral state: i.e., cells infected with wild-type SeV exhibited little or no increase in Stat1 levels and were permissive for VSV replication, even in the presence of exogenous IFN. In contrast, in cells infected with SeV lacking the AUG114-initiated C protein or containing a single amino acid substitution in the C protein, the level of Stat1 increased and VSV replication was inhibited. The prevention of the cellular IFN-mediated antiviral response appears to be a key determinant of SeV pathogenicity.


Assuntos
Antivirais/metabolismo , Interferon beta/metabolismo , Fosfoproteínas , Respirovirus , Proteínas Virais/fisiologia , Animais , Antivirais/farmacologia , Sítios de Ligação , Linhagem Celular , Cricetinae , Proteínas de Ligação a DNA/metabolismo , Interferon beta/farmacologia , Macaca mulatta , Camundongos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Respirovirus/genética , Respirovirus/metabolismo , Respirovirus/fisiologia , Fator de Transcrição STAT1 , Transativadores/metabolismo , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas Virais/genética , Proteínas Estruturais Virais/metabolismo
5.
J Virol ; 73(7): 5568-76, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364305

RESUMO

Paramyxoviruses cotranscriptionally edit their P gene mRNAs by expanding the number of Gs of a conserved AnGn run. Different viruses insert different distributions of guanylates, e.g., Sendai virus inserts a single G, whereas parainfluenza virus type 3 inserts one to six Gs. The sequences conserved at the editing site, as well as the experimental evidence, suggest that the insertions occur by a stuttering process, i.e., by pseudotemplated transcription. The number of times the polymerase "stutters" at the editing site before continuing strictly templated elongation is directed by a cis-acting sequence found upstream of the insertions. We have examined the stuttering process during natural virus infections by constructing recombinant Sendai viruses with mutations in their cis-acting sequences. We found that the template stutter site is precisely determined (C1052) and that a relatively short region (approximately 6 nucleotides) just upstream of the AnGn run can modulate the overall frequency of mRNA editing as well as the distribution of the nucleotide insertions. The positions more proximal to the 5' AnGn run are the most important in this respect. We also provide evidence that the stability of the mRNA/template hybrid plays a determining role in the overall frequency and range of mRNA editing. When the template U run is extended all the way to the stutter site, adenylates rather than guanylates are added at the editing site and their distribution begins to resemble the polyadenylation associated with mRNA 3' end formation by the viral polymerase. Our data suggest how paramyxovirus mRNA editing and polyadenylation are related mechanistically and how editing sites may have evolved from poly(A)-termination sites or vice versa.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Fosfoproteínas/metabolismo , Respirovirus/enzimologia , Respirovirus/genética , Proteínas Virais/metabolismo , Regiões 5' não Traduzidas , Animais , Embrião de Galinha , RNA Polimerases Dirigidas por DNA/genética , Fosfoproteínas/genética , Edição de RNA , RNA Mensageiro , RNA Viral , Células Tumorais Cultivadas , Proteínas Virais/genética
6.
J Virol ; 73(1): 343-51, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9847338

RESUMO

Editing of paramyxovirus P gene mRNAs occurs cotranscriptionally and functions to fuse an alternate downstream open reading frame to the N-terminal half of the P protein. G residues are inserted into a short G run contained within a larger purine run (AnGn) in this process, by a mechanism whereby the transcribing polymerase stutters (i.e., reads the same template cytosine more than once). Although Sendai virus (SeV) and bovine parainfluenza virus type 3 (bPIV3) are closely related, the G insertions in their P mRNAs are distributed differently. SeV predominantly inserts a single G residue within the G run of the sequence 5' AACAAAAAAGGG, whereas bPIV3 inserts one to six G's at roughly equal frequency within the sequence 5' AUUAAAAAAGGGG (differences are underlined). We have examined how the cis-acting editing sequence determines the number of G's inserted, both in a transfected cell system using minigenome analogues and by generating recombinant viruses. We found that the presence of four rather than three G's in the purine run did not affect the distribution of G insertions. However, when the underlined AC of the SeV sequence was replaced by the UU found in bPIV3, the editing phenotype from both the minigenome and the recombinant virus resembled that found in natural bPIV3 infections (i.e., a significant fraction of the mRNAs contained two to six G insertions). The two nucleotides located just upstream of the polypurine tract are thus key determinants of the editing phenotype of these viruses. Moreover, the minimum number of A residues that will promote SeV editing phenotype is six but can be reduced to five when the upstream AC is replaced by UU. A model for how the upstream dinucleotide controls the insertion phenotype is presented.


Assuntos
Edição de RNA , RNA Viral/genética , Respirovirus/genética , Sequência de Bases , RNA Polimerases Dirigidas por DNA/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Transfecção
7.
Mol Cell Biol ; 18(9): 5021-31, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9710586

RESUMO

The Sendai virus P/C mRNA expresses eight primary translation products by using a combination of ribosomal choice and cotranscriptional mRNA editing. The longest open reading frame (ORF) of the mRNA starts at AUG104 (the second initiation site) and encodes the 568-amino-acid P protein, an essential subunit of the viral polymerase. The first (ACG81), third (ATG114), fourth (ATG183), and fifth (ATG201) initiation sites are used to express a C-terminal nested set of polypeptides (collectively named the C proteins) in the +1 ORF relative to P, namely, C', C, Y1, and Y2, respectively. Leaky scanning accounts for translational initiation at the first three start sites (a non-ATG followed by ATGs in progressively stronger contexts). Consistent with this, changing ACG81/C' to ATG (GCCATG81G) abrogates expression from the downstream ATG104/P and ATG114/C initiation codons. However, expression of the Y1 and Y2 proteins remains normal in this background. We now have evidence that initiation from ATG183/Y1 and ATG201/Y2 takes place via a ribosomal shunt or discontinuous scanning. Scanning complexes appear to assemble at the 5' cap and then scan ca. 50 nucleotides (nt) of the 5' untranslated region before being translocated to an acceptor site at or close to the Y initiation codons. No specific donor site sequences are required, and translation of the Y proteins continues even when their start codons are changed to ACG. Curiously, ATG codons (in good contexts) in the P ORF, placed either 16 nt upstream of Y1, 29 nt downstream of Y2, or between the Y1 and Y2 codons, are not expressed even in the ACGY1/ACGY2 background. This indicates that ATG183/Y1 and ATG201/Y2 are privileged start sites within the acceptor site. Our observations suggest that the shunt delivers the scanning complex directly to the Y start codons.


Assuntos
Biossíntese de Proteínas , Respirovirus/genética , Respirovirus/metabolismo , Ribossomos/metabolismo , Proteínas Virais/biossíntese , Sequência de Bases , Linhagem Celular , Códon , Primers do DNA , Células HeLa , Humanos , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Fases de Leitura Aberta , Reação em Cadeia da Polimerase , Edição de RNA , RNA Mensageiro/metabolismo , RNA Viral/metabolismo
8.
J Virol ; 72(7): 5984-93, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9621061

RESUMO

Recombinant Sendai viruses were prepared which cannot express their Cprime, C, or Cprime plus C proteins due to mutation of their respective start codons ([Cprime-minus], [C-minus] and [double mutant], respectively). The [Cprime-minus] and [C-minus] stocks were similar to that of wild-type (wt) virus in virus titer and plaque formation, whereas the double-mutant stock had a much-reduced PFU or 50% egg infective dose/particle ratio and produced very small plaques. Relative to the wt virus infection, the [Cprime-minus] and [C-minus] infections of BHK cells resulted in significantly greater accumulation of viral RNAs, consistent with the known inhibitory effects of the Cprime and C proteins. The double-mutant infection, in contrast, was delayed in its accumulation of viral RNAs; however, once accumulation started, overaccumulation quickly occurred, as in the single-mutant infections. Our results suggest that the Cprime and C proteins both provide a common positive function early in infection, so that only the double mutant undergoes delayed RNA accumulation and exhibits the highly debilitated phenotype. Later in infection, the same proteins appear to act as inhibitors of RNA accumulation. In infections of mice, [Cprime-minus] was found to be as virulent as wt virus whereas [C-minus] was highly attenuated. These results suggest that the Cprime and C proteins cannot be functionally equivalent, since C can replace Cprime for virulence in mice whereas Cprime cannot replace C.


Assuntos
RNA Viral/biossíntese , Infecções por Respirovirus/virologia , Respirovirus/fisiologia , Proteínas não Estruturais Virais/fisiologia , Animais , Células Cultivadas , Cricetinae , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Recombinação Genética
9.
Virology ; 243(2): 340-53, 1998 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-9568033

RESUMO

The replication of nonsegmented minus-strand RNA genomes, like that of Sendai paramyxovirus (SeV), are controlled by the short leader regions present at each end of the linear genomes and antigenomes; the left and right promoters (PL and PR), respectively. Wild-type SeV is highly cytopathic in cell culture, because it induces programmed cell death (PCD). We have found that a recombinant SeV (rSeVGP42), in which the first 42 nt of le+ sequences at PL were replaced with the equivalent sequences of PR, and which produces infectious virus in amounts comparable to wild type, does not kill cells. Further, the increasing replacement of the terminal le+ sequences at PL with le- sequences led to a decreasing fraction of infected cells being apoptotic. This property (PCD-), moreover, is dominant in cells co-infected with SeVwt and rSeVGP42, and the mutant virus therefore appears to have gained a function which prevents PCD induced by SeVwt. Even though this virus has not been selected for naturally, it excludes SeVwt during co-infections of cultured cells or embryonated chicken eggs. The noncytopathic nature of cells infected or co-infected with rSeVGP42 leads automatically to stable, persistent infections. The mutation in rSeVGP42 is not in the protein coding regions of the viral genome, but in the 55-nt-long leader region which controls antigenome synthesis from genome templates. The SeV leader regions, which are expressed as short RNAs, thus appear to control the induction of PCD.


Assuntos
Apoptose , RNA Mensageiro , RNA Viral , Respirovirus/genética , Animais , Linhagem Celular , Mapeamento Cromossômico , Cricetinae , Efeito Citopatogênico Viral , Genoma Viral , Células HeLa , Humanos , Respirovirus/fisiologia , Latência Viral , Replicação Viral
10.
Virology ; 242(2): 327-37, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9514977

RESUMO

Wild-type Sendai virus expresses three proteins containing the N-terminal half of the P protein open reading frame due to mRNA editing; a full-length P protein (ca. 70% of the total), a V protein with the N-terminal half fused to a Cys-rich Zn(2+)-binding domain (ca. 25% of the total), and a W protein representing the N-terminal half alone (ca. 5% of the total). To examine the role of these proteins in the virus life cycle, we have prepared recombinant viruses in which the normal V mRNA expresses a W protein (V-stop; 70% P, 30% W), one which cannot edit its P gene mRNA (delta 6A; 100% P), and one which overedits its mRNA like parainfluenza virus type 3 (swap/8;20-40% P, 30% V, 30% W). All these viruses were readily recovered and grew to similar titers in eggs, and except for the P gene products, cell lines individually infected with these viruses accumulated similar amounts of viral macromolecules. The relative competitive advantage of each virus was determined by multiple cycle coinfections of eggs and found to be rSeV-Vstop = rSeV-wt >> rSeV-delta 6A > rSeV-swap/8. On the other hand, rSeV-swap/8 underwent multiple cycles of replication in C57BI/6 mouse lungs and was highly virulent for these animals, whereas rSeV-delta 6A was avirulent in mice and this infection was quickly cleared. Remarkably, rSeV-Vstop appeared to be more virulent for inbred C57BI/6 mice than rSeV-wt, but was partially attenuated in infections of outbred ICR mice. Thus, the expression of either the V or the W proteins is sufficient for multiple cycles of infection and pathogenesis in C57BI/6 mice, whereas W can only partially substitute for V for pathogenesis in ICR mice.


Assuntos
Fosfoproteínas/metabolismo , Respirovirus/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/fisiologia , Animais , Peso Corporal , Células Cultivadas , Embrião de Galinha , Cricetinae , Cães , Expressão Gênica , Haplorrinos , Humanos , Pneumopatias/patologia , Pneumopatias/virologia , Camundongos , Camundongos Endogâmicos C57BL/virologia , Camundongos Endogâmicos ICR/virologia , RNA Viral/análise , Proteínas Recombinantes/metabolismo , Respirovirus/patogenicidade
12.
Virology ; 238(2): 424-31, 1997 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-9400614

RESUMO

A mutant Sendai virus (SevMVC), which grows much better than its progenitor virus (SeVM) in cell culture, but, in strong contrast to SeVM, is totally avirulent for mice, has been described. SeVMVC contains two amino acid substitutions relative to SeVM, namely, F170S in the C protein and E2050A in the L protein. We have examined which substitutions were responsible for the above phenotypes by exchanging the C gene of our reference strain Z with those of SeVH (another reference strain), SeVM, and SeVMVC, in turn. We have found that the F170S mutation in the CMVC protein is responsible both for enhanced replication in cell culture and for avirulence in mice. Avirulence appeared to be due to restricted viral replication primarily after day 1, implicating some aspect of innate immunity in this process. The SeV C proteins thus appear to be required for multiple cycles of replication in mice.


Assuntos
Respirovirus/patogenicidade , Proteínas Virais/fisiologia , Animais , Células Cultivadas , Camundongos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Mutação Puntual , Respirovirus/genética , Respirovirus/crescimento & desenvolvimento , Proteínas Virais/genética , Proteínas Virais/metabolismo , Virulência
13.
J Virol ; 71(12): 9588-99, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9371623

RESUMO

The role of the negative-stranded virus accessory C proteins is difficult to assess because they appear sometimes as nonessential and thereby of no function. On the other hand, when a function is found, as in the case of Sendai virus, it represents an enigma, in that the C proteins inhibit replication under conditions where the infection follows an exponential course. Furthermore, this inhibitory function is exerted differentially: in contrast to the replication of internal deletion defective interfering (DI) RNAs, that of copy-back DI RNAs appears to escape inhibition, under certain experimental conditions (in vivo assay). In a reexamination of the C effect by the reverse genetics approach, it was found that copy-back RNA replication is inhibited by C in vivo as well, under conditions where the ratio of C to copy-back template is increased. This effect can be reversed by an increase in P but not L protein. The "rule of six" was differentially observed in the presence or absence of C. Finally, a difference in the ability of the replicating complex to tolerate promoter modifications in RNA synthesis initiation was shown to occur in the presence or the absence of C as well. We propose that C acts by increasing the selectivity of the replicating complex for the promoter cis-acting elements governing its activity. The inhibitory effect of C becomes the price to pay for this increased selectivity.


Assuntos
RNA Polimerases Dirigidas por DNA , Regiões Promotoras Genéticas , RNA Viral/biossíntese , Respirovirus/genética , Proteínas Virais/metabolismo , Replicação Viral , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Genoma Viral , Células HeLa , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Respirovirus/fisiologia , Proteínas Virais/genética
14.
Virology ; 228(1): 55-62, 1997 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-9024809

RESUMO

The Sendai virus V protein is a nonstructural trans-frame protein in which a highly conserved cys-rich Zn2+-binding domain is fused to the N-terminal half of the P protein via mRNA editing. Using a recently developed system in which infectious virus is recovered from cDNA, we have engineered a virus in which a translation stop codon was placed at the beginning of the V ORF. Translation of the V(stop) mRNA yields a W-like protein, i.e., a protein composed of the N-terminal half of the P protein alone which is naturally expressed at low levels from the P gene. This V-minus but W-augmented virus was found to replicate normally in cell culture and embryonated chicken eggs. The Sendai virus V protein is thus an accessory protein, and the cys-rich Zn2+-binding domain is likely to function in a specialized role during virus propagation.


Assuntos
Respirovirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Embrião de Galinha , Cricetinae , Cães , Expressão Gênica , Células HeLa , Humanos , Macaca mulatta , Fases de Leitura Aberta , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Respirovirus/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/isolamento & purificação
15.
Virology ; 224(2): 405-14, 1996 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8874501

RESUMO

We have used a cDNA copy of a natural, internally deleted, Sendai virus defective interfering genome to study the effect of insertions and deletions (which maintain the hexamer genome length) on the ability of viral genomes to be amplified in a transfected cell system. The insertion of 18 nt at nt72 (In the 5' untranslated region of the N gene, just downstream of the le+ region) was found to be lethal, whereas similar insertions further from the genome ends were well tolerated. Curiously, the insertion of 6 nt on either side of the le+/N junction (at nt47 and nt87) was well tolerated, but the insertion of 12 nt at either site, or of 6 nt at both sites, largely ablated genome amplification. These results suggest that an element of this replication promoter is located downstream of nt87, in the 5' untranslated region of the first gene. Remarkably, the addition of 6 nt by the insertion of 2, 3, or 4 nt at nt47 plus the insertion of 4, 3, or 2 nt, respectively, at nt87 was poorly tolerated, presumably because the hexamer phase of the intervening sequence was altered with respect to the N subunits of the template. These results suggest that the rule of six operates, at least in part, at the level of the initiation of antigenome synthesis.


Assuntos
Regiões Promotoras Genéticas , Respirovirus/genética , Replicação Viral/genética , Genoma Viral , Humanos , Mutagênese Insercional , Respirovirus/fisiologia , Deleção de Sequência , Células Tumorais Cultivadas
16.
RNA ; 2(10): 1033-45, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8849779

RESUMO

Paramyxoviruses cotranscriptionally edit their P gene mRNA by the programmed insertion of G residues into a short G run contained within a larger purine run, via pseudo-templated transcription. The templates for paramyxovirus transcription are genome nucleocapsids in which each nucleoprotein subunit is associated with 6 nt, and only genomes whose lengths are multiples of 6 are found naturally or are replicated efficiently in transfected cell systems. We have examined the effect of varying total genome length on the frequency and number of insertions into the mRNA editing site in a transfected cell system, using constructs that generate mini-genome analogues. We found that, as long as the purine run sequence and the region immediately upstream were unaltered, editing occurred during mRNA synthesis independent of the precise length of the minigenome. However, when mini-genome constructs whose lengths were not multiples of 6 were used, insertions (or deletions) occurred during antigenome synthesis within the purine run, which strikingly restored the hexamer length. Genome length correction due to changes in the antigenome purine run length occurred only when the mini-genome was not a multiple of 6, and these changes were only poorly affected by mutations in the mRNA editing site and the region immediately upstream. Our results suggest that the mRNA editing site is a natural hotspot for viral polymerase slippage during genome replication, and that this site serves the dual and complementary function of maintaining hexamer genome length. The unusual requirement of paramyxoviruses for genomes of precise hexamer length may have evolved to maintain genome stability against insertions in the mRNA editing site during replication.


Assuntos
Genoma Viral , Fosfoproteínas , Edição de RNA/genética , RNA Viral/metabolismo , Respirovirus/genética , Proteínas Virais/genética , Composição de Bases , RNA Polimerases Dirigidas por DNA/metabolismo , Guanidina , Guanidinas , Peso Molecular , Mutação , Fenótipo , Nucleotídeos de Purina , RNA Mensageiro/biossíntese , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/biossíntese , RNA Viral/química , RNA Viral/genética , Replicação Viral/genética
17.
J Virol ; 70(8): 5067-74, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8764014

RESUMO

Many paramyxoviruses express small basic C proteins, from an alternate, overlapping open reading frame of the P gene mRNA, which were previously found to inhibit mRNA synthesis. During recent experiments in which infectious Sendai virus (SeV) was recovered from cDNA via the initial expression of the viral N, P, and L genes from plasmids, the abrogation of C protein expression from the plasmid P gene was found to be necessary for virus recovery. We have investigated the effect of C coexpression on the amplification of an internally deleted defective interfering (DI) genome directly in the transfected cell, for which, in contrast to virus recovery experiments, genome amplification is independent of mRNA synthesis carried out by the SeV polymerase. We find that C protein coexpression also strongly inhibits the amplification of this DI genome but has little or no effect on that of a copy-back DI genome (DI-H4). We have also characterized the C protein from a mutant SeV and found that (i) it had lost most of its inhibitory activity on internally deleted DI genome amplification and (ii) its coexpression no longer prevented the recovery of SeV from DNA. However, consistent with the insensitivity of copy-back DI genomes to C protein inhibition, C coexpression did not prevent the recovery of copy-back nondefective viruses from DNA. The inhibitory effects of C coexpression thus appear to be promoter specific.


Assuntos
Amplificação de Genes , Genoma Viral , Respirovirus/genética , Proteínas Virais/genética , Regulação da Expressão Gênica , Mutação , Regiões Promotoras Genéticas/genética , Respirovirus/metabolismo
18.
Virology ; 214(1): 139-49, 1995 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8525609

RESUMO

When HA epitope-tagged and untagged Sendai virus (SeV) P proteins are coexpressed and the products reacted with anti-HA, the untagged P protein is also selected because this protein is found as an oligomer. The oligomer was determined to be a homotrimer by coselection studies in which increasing amounts of untagged versus tagged protein were coexpressed, and these findings were extended to mumps virus, a member of the rubulavirus genus. The region of the SeV protein responsible for the oligomerization was localized to residues 344-411. Computer analysis of the 13 Paramyxovirus P proteins in the database revealed that all but one are predicted to form coiled coils in this region, the first of only two regions that can be aligned throughout the entire virus subfamily. The predicted coiled-coil region of the measles virus P protein, when grafted onto the C-terminus of the normally monomeric La protein, led to the efficient oligomerization of this reporter protein. The predicted coiled-coil region of these P proteins thus appears to be sufficient for oligomerization.


Assuntos
Vírus da Parainfluenza 1 Humana/metabolismo , Fosfoproteínas/química , Proteínas Virais/química , Sequência de Aminoácidos , Animais , Linhagem Celular Transformada , Epitopos , Células HeLa , Humanos , Dados de Sequência Molecular , Fosfoproteínas/metabolismo , Conformação Proteica , Homologia de Sequência de Aminoácidos , Transfecção , Proteínas Virais/metabolismo
19.
EMBO J ; 14(24): 6087-94, 1995 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8557028

RESUMO

We have recovered infectious Sendai virus (SeV) from full-length cDNA (FL-3) by transfecting this cDNA and pGEM plasmids expressing the nucleocapsid protein (NP), phosphoprotein and large proteins into cells infected with a vaccinia virus which expresses T7 RNA polymerase. These cells were then injected into chicken eggs, in which SeV grows to very high titers. FL-3 was marked with a BglII site in the leader region and an NsiI site (ATGCAT) in the 5' nontranslated region of the NP gene, creating a new, out-of-frame, 5' proximal AUG. All the virus stocks generated eventually removed this impediment to NP expression, by either point mutation or recombination between FL-3 and pGEM-NP. The recovery system was found to be highly recombinogenic. Even in the absence of selective pressure, one in 20 of the recombinant SeV generated had exchanged the NP gene of FL-3 with that of pGEM-NP. When a fifth plasmid containing a new genomic 3' end without the presumably deleterious BglII site was included as another target for recombination, the new genomic 3' end was found in the recombinant SeV in 12 out of 12 recoveries. Using this approach, a novel copy-back nondefective virus was generated which interferes with wild-type virus replication.


Assuntos
DNA Complementar/genética , DNA Viral/genética , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 1 Humana/isolamento & purificação , Animais , Sequência de Bases , Linhagem Celular , Galinhas , Primers do DNA/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 1 Humana/crescimento & desenvolvimento , Plasmídeos/genética , Mutação Puntual , Reação em Cadeia da Polimerase , RNA Viral/genética , Recombinação Genética , Transfecção , Vaccinia virus/genética , Proteínas Virais/genética
20.
J Virol ; 69(9): 5754-62, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7637020

RESUMO

We examined the 5' ends of Hantaan virus (HTN) genomes and mRNAs to gain insight into the manner in which these chains were initiated. Like those of all members of the family Bunyaviridae described so far, the HTN mRNAs contained 5' terminal extensions that were heterogeneous in both length and sequence, presumably because HTN also "cap snatches" host mRNAs to initiate the viral mRNAs. Unexpectedly, however, almost all of the mRNAs contained a G residue at position -1, and a large fraction also lacked precisely one of the three UAG repeats at the termini. The genomes, on the other hand, commenced with a U residue at position +1, but only 5' monophosphates were found here, indicating that these chains may not have initiated with UTP at this position. Taken together, these unusual findings suggest a prime-and-realign mechanism of chain initiation in which mRNAs are initiated with a G-terminated host cell primer and genomes with GTP, not at the 3' end of the genome template but internally (opposite the template C at position +3), and after extension by one or a few nucleotides, the nascent chain realigns backwards by virtue of the terminal sequence repeats, before processive elongation takes place. For genome initiation, an endonuclease, perhaps that involved in cap snatching, is postulated to remove the 5' terminal extension of the genome, leaving the 5' pU at position +1.


Assuntos
Bunyaviridae/genética , Vírus Hantaan/genética , RNA Mensageiro/biossíntese , RNA Viral/biossíntese , RNA Viral/genética , Animais , Sequência de Bases , Bunyaviridae/metabolismo , Primers do DNA , DNA Complementar , Genoma Viral , Guanosina Trifosfato/metabolismo , Vírus Hantaan/metabolismo , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Capuzes de RNA/biossíntese , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Viral/química , Sequências Repetitivas de Ácido Nucleico , Especificidade da Espécie , Moldes Genéticos , Transcrição Gênica , Uridina Trifosfato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...