Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Parasite Immunol ; 37(9): 433-45, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26121587

RESUMO

Plasmodium falciparum infections have been implicated in immune deficiencies resulting in ineffective control of Epstein-Barr virus, thereby increasing the risk of endemic Burkitt lymphoma in children. However, the impact of Epstein-Barr virus infections on the development of immunity to P. falciparum has not been studied in depth. In this review, we examine novel findings from animal co-infection models and human immuno-epidemiologic studies to speculate on the impact of acute gammaherpesvirus co-infection on malarial disease severity. Children are often concurrently or sequentially infected with multiple pathogens, and this has implications for understanding the development of protective immunity as well as in the evaluation of vaccine efficacy.


Assuntos
Coinfecção/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Herpesvirus Humano 4/fisiologia , Malária Falciparum/imunologia , Doença Aguda , África Subsaariana/epidemiologia , Animais , Linfoma de Burkitt/parasitologia , Linfoma de Burkitt/virologia , Criança , Citocinas/imunologia , Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/epidemiologia , Humanos , Malária Falciparum/epidemiologia , Linfócitos T/imunologia
2.
Am J Transplant ; 15(10): 2576-87, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25989700

RESUMO

Latent viral infections are a major concern among immunosuppressed transplant patients. During clinical trials with belatacept, a CTLA4-Ig fusion protein, patients showed an increased risk of Epstein-Barr virus-associated posttransplant lymphoproliferative disorder, thought to be due to a deficient primary CD8(+) T cell response to the virus. Using a murine model of latent viral infection, we observed that rapamycin treatment alone led to a significant increase in virus-specific CD8(+) T cells, as well as increased functionality of these cells, including the ability to make multiple cytokines, while CTLA4-Ig treatment alone significantly dampened the response and inhibited the generation of polyfunctional antigen-specific CD8(+) T cells. However, the addition of rapamycin to the CTLA4-Ig regimen was able to quantitatively and qualitatively restore the antigen-specific CD8(+) T cell response to the virus. This improvement was physiologically relevant, in that CTLA4-Ig treated animals exhibited a greater viral burden following infection that was reduced to levels observed in untreated immunocompetent animals by the addition of rapamycin. These results reveal that modulation of T cell differentiation though inhibition of mTOR signaling can restore virus-specific immune competence even in the absence of CD28 costimulation, and have implications for improving protective immunity in transplant recipients.


Assuntos
Abatacepte/efeitos adversos , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/imunologia , Gammaherpesvirinae , Infecções por Herpesviridae/tratamento farmacológico , Imunossupressores/uso terapêutico , Sirolimo/uso terapêutico , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Quimioterapia Combinada , Infecções por Herpesviridae/imunologia , Imunossupressores/efeitos adversos , Imunossupressores/farmacologia , Camundongos , Sirolimo/farmacologia
3.
Science ; 345(6196): 573-7, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24968940

RESUMO

Mammals are coinfected by multiple pathogens that interact through unknown mechanisms. We found that helminth infection, characterized by the induction of the cytokine interleukin-4 (IL-4) and the activation of the transcription factor Stat6, reactivated murine γ-herpesvirus infection in vivo. IL-4 promoted viral replication and blocked the antiviral effects of interferon-γ (IFNγ) by inducing Stat6 binding to the promoter for an important viral transcriptional transactivator. IL-4 also reactivated human Kaposi's sarcoma-associated herpesvirus from latency in cultured cells. Exogenous IL-4 plus blockade of IFNγ reactivated latent murine γ-herpesvirus infection in vivo, suggesting a "two-signal" model for viral reactivation. Thus, chronic herpesvirus infection, a component of the mammalian virome, is regulated by the counterpoised actions of multiple cytokines on viral promoters that have evolved to sense host immune status.


Assuntos
Gammaherpesvirinae/fisiologia , Herpesvirus Humano 8/fisiologia , Interferon gama/imunologia , Interleucina-4/metabolismo , Fator de Transcrição STAT6/metabolismo , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Ativação Viral/fisiologia , Animais , Gammaherpesvirinae/genética , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Humanos , Interferon gama/farmacologia , Interleucina-4/farmacologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Nematospiroides dubius/imunologia , Óvulo/imunologia , Regiões Promotoras Genéticas , Infecções por Strongylida/imunologia , Ativação Viral/efeitos dos fármacos , Ativação Viral/genética , Latência Viral/fisiologia , Replicação Viral/fisiologia
4.
J Clin Invest ; 107(2): R15-22, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11160143

RESUMO

Infection of medial smooth muscle cells with gamma-herpesvirus 68 (gammaHV68) causes severe chronic vasculitis that is restricted to the great elastic arteries. We show here that persistence of disease in the great elastic arteries is (a) due to inefficient clearance of viral infection from this site compared with other organs or other vascular sites, and (b) associated with failure of T cells and macrophages to enter the virus-infected elastic media. These findings demonstrate immunoprivilege of the media of the great elastic arteries. We found that IFN-gamma acted on somatic cells during acute infection to prevent the establishment of medial infection and on hematopoietic cells to determine the severity of disease in this site. The immunoprivileged elastic media may provide a site for persistence of pathogens or self antigens leading to chronic vascular disease, a process regulated by IFN-gamma actions on both somatic and hematopoietic cells. These concepts have significant implications for understanding immune responses contributing to or controlling chronic inflammatory diseases of the great vessels.


Assuntos
Aorta/efeitos dos fármacos , Aortite/virologia , Herpes Simples/imunologia , Interferon gama/farmacologia , Simplexvirus , Animais , Antígenos Virais/análise , Aorta/virologia , Aortite/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Doença Crônica , Fígado/virologia , Pulmão/virologia , Camundongos , Baço/virologia , Fatores de Tempo , Tropismo , beta-Galactosidase/análise
5.
J Virol ; 74(23): 11115-20, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11070007

RESUMO

Epstein-Barr virus (EBV) nuclear antigen (EBNA) 2 (EBNA2) is involved in upregulating the expression of both EBNAs and latency-associated membrane proteins. Transcription of the six EBNA genes, which are expressed in EBV-immortalized primary B cells, arises from one of two promoters, Cp and Wp, located near the left end of the viral genome. Wp is exclusively used to drive EBNA gene transcription during the initial stages of infection in primary B cells; induction of transcription from Cp follows. We previously have mapped an EBNA2-dependent enhancer upstream of Cp (M. Woisetschlaeger et al., Proc. Natl. Acad. Sci. USA 88:3942-3946, 1991) and, more recently, have demonstrated that deletion of this enhancer results in EBV-immortalized lymphoblastoid cell lines (LCLs) that are heavily biased toward the use of Wp to drive transcription of the EBNA genes (L. Yoo et al., J. Virol. 71:9134-9142, 1997). To assess the immortalizing capacity of this mutant EBV and to monitor the early events after infection of primary B cells, B cells isolated from cottontop marmosets were used to generate LCLs immortalized with the Cp EBNA2 enhancer deletion mutant virus. As previously reported, all EBV-infected marmoset LCLs examined could be triggered to produce significant levels of virus. Infection of human B cells with wild-type or Cp EBNA2 enhancer mutant viruses recovered from marmoset B-cell lines demonstrated that (i) the Cp EBNA2 enhancer mutant virus immortalizes primary human B cells nearly as efficiently as wild-type virus and (ii) the Cp EBNA2-dependent enhancer plays an important role in the induction of Cp activity during the early stages of infection. The latter is consistent with the phenotype of LCLs immortalized with the Cp EBNA2 enhancer mutant EBV. Finally, using an established LCL in which EBNA2 function is regulated by beta-estradiol, we showed that the loss of EBNA2 function results in an approximately 4-fold decrease in the steady-state levels of Cp-initiated transcripts and a concomitant increase in the steady-state levels of Wp-initiated transcripts. Taken together, these results provide strong evidence that EBNA2 plays an important role in regulating Cp activity. These results also demonstrate that diminished induction of Cp activity does not appear to affect the ability of EBV to immortalize primary B cells in cultures. Finally, as shown here, infection of marmoset B cells with immortalization-competent mutants of EBV provides a convenient reservoir for the production of mutant viruses.


Assuntos
Elementos Facilitadores Genéticos , Antígenos Nucleares do Vírus Epstein-Barr/fisiologia , Herpesvirus Humano 4/genética , Regiões Promotoras Genéticas , Latência Viral , Animais , Callithrix , Linhagem Celular , Herpesvirus Humano 4/fisiologia , Humanos , Mutação
6.
J Virol ; 74(23): 11304-10, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11070030

RESUMO

The role of autoimmunity in large-vessel vasculitis in humans remains unclear. We have previously shown that infection of gamma interferon receptor knockout (IFN-gamma R(-/-)) mice with gammaherpesvirus 68 (gamma HV68) results in severe inflammation of the large elastic arteries that is pathologically similar to the lesions observed in Takayasu's arteritis, the nongranulomatous variant of temporal arteritis, and Kawasaki's disease (K. E. Weck et al., Nat. Med. 3:1346-1353, 1997). Here we define the mechanism of damage to the elastic arteries. We show that there is a persistent productive infection of the media of the large elastic vessels. In addition, we demonstrate that persistent virus replication is necessary for chronic arteritis, since antiviral therapy of mice with established disease resulted in increased survival, clearance of viral antigen from the media of the affected vessel, and dramatic amelioration of arteritic lesions. These data argue that ongoing virus replication, rather than autoimmunity, is the cause of gamma HV68-induced elastic arteritis.


Assuntos
Arterite/etiologia , Gammaherpesvirinae/patogenicidade , Organofosfonatos , Replicação Viral , Animais , Células Apresentadoras de Antígenos/fisiologia , Aorta/virologia , Autoimunidade , Cidofovir , Citosina/análogos & derivados , Citosina/farmacologia , Camundongos , Camundongos SCID , Compostos Organofosforados/farmacologia , Receptores de Interferon/fisiologia , Replicação Viral/efeitos dos fármacos , Receptor de Interferon gama
7.
J Virol ; 74(15): 6741-7, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10888612

RESUMO

Chemokines are involved in recruitment and activation of hematopoietic cells at sites of infection and inflammation. The M3 gene of gammaHV68, a gamma-2 herpesvirus that infects and establishes a lifelong latent infection and chronic vasculitis in mice, encodes an abundant secreted protein during productive infection. The M3 gene is located in a region of the genome that is transcribed during latency. We report here that the M3 protein is a high-affinity broad-spectrum chemokine scavenger. The M3 protein bound the CC chemokines human regulated upon activation of normal T-cell expressed and secreted (RANTES), murine macrophage inflammatory protein 1alpha (MIP-1alpha), and murine monocyte chemoattractant protein 1 (MCP-1), as well as the human CXC chemokine interleukin-8, the murine C chemokine lymphotactin, and the murine CX(3)C chemokine fractalkine with high affinity (K(d) = 1. 6 to 18.7 nM). M3 protein chemokine binding was selective, since the protein did not bind seven other CXC chemokines (K(d) > 1 microM). Furthermore, the M3 protein abolished calcium signaling in response to murine MIP-1alpha and murine MCP-1 and not to murine KC or human stromal cell-derived factor 1 (SDF-1), consistent with the binding data. The M3 protein was also capable of blocking the function of human CC and CXC chemokines, indicating the potential for therapeutic applications. Since the M3 protein lacks homology to known chemokines, chemokine receptors, or chemokine binding proteins, these studies suggest a novel herpesvirus mechanism of immune evasion.


Assuntos
Quimiocinas CX3C , Quimiocinas C , Quimiocinas/metabolismo , Gammaherpesvirinae/metabolismo , Proteínas Virais/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Quimiocina CX3CL1 , Quimiocinas CC/metabolismo , Quimiocinas CXC/metabolismo , Gammaherpesvirinae/genética , Humanos , Linfocinas/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Sialoglicoproteínas/metabolismo , Proteínas Virais/genética , Proteínas Virais/farmacologia
8.
J Virol ; 74(16): 7451-61, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10906198

RESUMO

Gamma-2 herpesviruses encode a homolog of mammalian D-type cyclins. The v-cyclin encoded by murine gammaherpesvirus 68 (gammaHV68) induces cell cycle progression and is an oncogene (L. F. van Dyk, J. L. Hess, J. D. Katz, M. Jacoby, S. H. Speck, and H. W. Virgin IV, J. Virol. 73:5110-5122, 1999). However, the role of the pro-proliferative v-cyclins in gamma-2 herpesvirus pathogenesis is not known. Here we report the generation and characterization of a gammaHV68 v-cyclin mutant (v-cyclin.LacZ) that is unable to express a functional v-cyclin protein. Notably, although the gammaHV68 v-cyclin is expressed from an early-late lytic transcript, v-cyclin. LacZ replicated normally in fibroblasts in vitro and during acute infection in the spleen, liver, and lungs in vivo. Moreover, v-cyclin.LacZ exhibited wild-type (wt) virulence in mice with severe combined immunodeficiency. In addition, in a model of gammaHV68-induced chronic disease in mice lacking the gamma interferon receptor (IFNgammaR(-/-)), v-cyclin.LacZ virus was similar to wt gammaHV68 in terms of the incidence of mortality and vasculitis. Further analysis revealed that the frequencies of splenocytes and peritoneal cells harboring the latent gammaHV68 genome in normal and B-cell-deficient mice infected with wt gammaHV68 or v-cyclin.LacZ were very similar. However, v-cyclin.LacZ was significantly compromised in its capacity to reactivate from latency. This phenotype was conclusively mapped to the v-cyclin gene by (i) generating a marker rescue virus (v-cyclin.MR) from the v-cyclin.LacZ mutant, which restored the frequency of cells in which virus reactivated from latency to the levels observed with wt gammaHV68; and (ii) generating a second v-cyclin mutant virus containing a translation stop codon within the v-cyclin gene (v-cyclin.stop), which was compromised in reactivation from latency. These studies demonstrate that despite expression as a lytic cycle gene, the pro-proliferative gammaHV68 v-cyclin is not required for gammaHV68 replication either in vitro or during acute infection in vivo but rather is a critical determinant of reactivation from latency.


Assuntos
Ciclinas/fisiologia , Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/virologia , Ativação Viral , Latência Viral , Animais , Células Cultivadas , Ciclinas/genética , Fibroblastos , Gammaherpesvirinae/genética , Gammaherpesvirinae/patogenicidade , Regulação Viral da Expressão Gênica , Marcação de Genes , Interferon gama/deficiência , Interferon gama/fisiologia , Camundongos , Camundongos SCID , Proteínas Virais , Virulência , Replicação Viral
9.
Nat Med ; 6(3): 343-5, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10700239

RESUMO

The hematopoietic-specific transmembrane protein tyrosine phosphatase CD45 functions to regulate Src kinases required for T- and B-cell antigen receptor signal transduction. So far, there have been no reports to our knowledge of a human deficiency in a tyrosine-specific phosphatase. Here, we identified a male patient with a deficiency in CD45 due to a large deletion at one allele and a point mutation at the other. The point mutation resulted in the alteration of intervening sequence 13 donor splice site. The patient presented at 2 months of age with severe combined immunodeficiency disease. The population of peripheral blood T lymphocytes was greatly diminished and unresponsive to mitogen stimulation. Despite normal B-lymphocyte numbers, serum immunoglobulin levels decreased with age. Thus, CD45 deficiency in humans results in T- and B-lymphocyte dysfunction.


Assuntos
Antígenos CD/genética , Linfócitos B/imunologia , Antígenos Comuns de Leucócito/genética , Deleção de Sequência , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Linfócitos T/imunologia , Antígenos CD/sangue , Sequência de Bases , Éxons , Feminino , Humanos , Imunoglobulina M/sangue , Lactente , Células Matadoras Naturais/imunologia , Antígenos Comuns de Leucócito/sangue , Contagem de Linfócitos , Masculino , Dados de Sequência Molecular , Linhagem , Mapeamento por Restrição , Imunodeficiência Combinada Severa/terapia
10.
J Virol ; 74(4): 1973-84, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10644370

RESUMO

Murine gammaherpesvirus 68 (gammaHV68, or MHV-68) is a genetically tractable, small animal model for the analysis of gammaherpesvirus pathogenesis. The gammaHV68 genome is colinear with the genomes of other sequence gammaherpesviruses, containing large blocks of conserved genes interspersed by a number of putative genes without clear homologs in the other gammaherpesviruses. One of these putative unique genes, the M1 open reading frame (ORF), exhibits sequence homology to a poxvirus serine protease inhibitor, SPI-1, as well as to another gammaHV68 gene, M3, which we have recently shown encodes an abundantly secreted chemokine binding protein. To assess the contribution of the M1 ORF to gammaHV68 pathogenesis, we have generated a recombinant gammaHV68 in which the M1 ORF has been disrupted through targeted insertion of a lacZ expression cassette (M1.LacZ). Although M1.LacZ replicated normally in tissue culture, it exhibited decreased splenic titers at days 4 and 9 postinfection in both immunocompetent and immunodeficient mice. Despite decreased levels of acute virus replication, M1.LacZ established a latent infection comparable to wild-type (wt) gammaHV68, but exhibited an approximately fivefold increase in efficiency of reactivation from latency. M1.LacZ also caused severe vasculitis of the great elastic arteries in gamma interferon receptor (IFN-gammaR)-deficient mice with a frequency comparable to wt gammaHV68, but did not cause the mortality or splenic pathology observed with wt gammaHV68 infection of IFN-gammaR-deficient mice. Restoration of M1 ORF sequences into M1.LacZ (M1 marker rescue, or M1.MR) demonstrated that M1.LacZ phenotypic alterations in growth in vivo and latency were not due to the presence of additional mutations located elsewhere in the M1. LacZ genome. Generation of a second M1 mutant virus containing a deletion at the 5' end of the M1 ORF (M1Delta511), but lacking the LacZ expression cassette, revealed the same latency phenotype observed with the M1.LacZ mutant. However, M1Delta511 was not attenuated for acute virus replication in the spleen. We conclude that (i) the induction of arteritis in gammaHV68-infected IFN-gammaR-deficient mice can occur in the absence of splenic pathology and mortality, (ii) replication during acute infection is not the primary determinant for the establishment of latent infection, and (iii) the M1 ORF, or a closely linked gene, encodes a gene product that functions to suppress virus reactivation.


Assuntos
Gammaherpesvirinae/crescimento & desenvolvimento , Fases de Leitura Aberta , Ativação Viral , Animais , Linhagem Celular , Gammaherpesvirinae/genética , Gammaherpesvirinae/patogenicidade , Gammaherpesvirinae/fisiologia , Marcação de Genes , Imunocompetência , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Baço/virologia , Virulência , Latência Viral , Replicação Viral
11.
J Virol ; 74(4): 2029-37, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10644377

RESUMO

Gene 50 is the only immediate-early gene that appears to be conserved among the characterized gammaherpesviruses. It has recently been demonstrated for the human viruses Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) that ectopic expression of the gene 50-encoded product in some latently infected cell lines can lead to the induction of virus replication, indicating that gene 50 is likely to play a pivotal role in regulating gammaherpesvirus reactivation. Here we demonstrate that the murine gammaherpesvirus 68 (gammaHV68) gene 50 is an immediate-early gene and that transcription of gammaHV68 gene 50 leads to the production of both spliced and unspliced forms of the gene 50 transcript. Splicing of the transcript near the 5' end serves to extend the gene 50 open reading frame, as has been observed for the gene 50 transcripts encoded by KSHV and herpesvirus saimiri (Whitehouse et al., J. Virol. 71:2550-2554, 1997; Lukac et al., Virology 252:304-312, 1998; Sun et al., Proc. Natl. Acad. Sci. USA 95:10866-10871, 1998). Reverse transcription-PCR analyses, coupled with S1 nuclease protection assays, provided evidence that gene 50 transcripts initiate at several sites within the region from bp 66468 to 66502 in the gammaHV68 genome. Functional characterization of the region upstream of the putative gene 50 transcription initiation site demonstrated orientation-dependent promoter activity and identified a 110-bp region (bp 66442 to 66552) encoding the putative gene 50 promoter. Finally, we demonstrate that the gammaHV68 gene 50 can transactivate the gammaHV68 gene 57 promoter, a known early gene target of the gene 50-encoded transactivator in other gammaherpesviruses. These studies show that the gammaHV68 gene 50 shares several important molecular similarities with the gene 50 homologs in other gammaherpesviruses and thus provides an impetus for future studies analyzing the role of the gammaHV68 gene 50-encoded protein in acute virus replication and reactivation from latency in vivo.


Assuntos
Gammaherpesvirinae/genética , Regulação Viral da Expressão Gênica , Proteínas Imediatamente Precoces/genética , Transativadores/genética , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Mapeamento Cromossômico , DNA Viral , Humanos , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Splicing de RNA , RNA Viral , Homologia de Sequência de Aminoácidos , Transativadores/metabolismo , Transcrição Gênica , Ativação Transcricional , Proteínas Virais/metabolismo
12.
J Virol ; 73(12): 9959-68, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10559309

RESUMO

Epstein-Barr virus (EBV) establishes a life-long infection in humans, with distinct viral latency programs predominating during acute and chronic phases of infection. Only a subset of the EBV latency-associated antigens present during the acute phase of EBV infection are expressed in the latently infected memory B cells that serve as the long-term EBV reservoir. Since the EBV immortalization program elicits a potent cellular immune response, downregulation of viral gene expression in the long-term latency reservoir is likely to facilitate evasion of the immune response and persistence of EBV in the immunocompetent host. Tissue culture and tumor models of restricted EBV latency have consistently demonstrated a critical role for methylation of the viral genome in maintaining the restricted pattern of latency-associated gene expression. Here we extend these observations to demonstrate that the EBV genomes in the memory B-cell reservoir are also heavily and discretely methylated. This analysis reveals that methylation of the viral genome is a normal aspect of EBV infection in healthy immunocompetent individuals and is not restricted to the development of EBV-associated tumors. In addition, the pattern of methylation very likely accounts for the observed inhibition of the EBV immortalization program and the establishment and maintenance of a restricted latency program. Thus, EBV appears to be the first example of a parasite that usurps the host cell-directed methylation system to regulate pathogen gene expression and thereby establish a chronic infection.


Assuntos
Metilação de DNA , Antígenos Nucleares do Vírus Epstein-Barr/genética , Infecções por Herpesviridae/virologia , Herpesvirus Humano 4/genética , Regiões Promotoras Genéticas , Infecções Tumorais por Vírus/virologia , Latência Viral , DNA Viral/metabolismo , Nível de Saúde , Humanos
13.
J Virol ; 73(9): 7658-70, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10438856

RESUMO

Sequence analysis of the murine gammaherpesvirus 68 (gammaHV68) genome revealed an open reading frame (gene 4) which is homologous to a family of proteins known as the regulators of complement activation (RCA proteins) (H. W. Virgin, P. Latreille, P. Wamsley, K. Hallsworth, K. E. Weck, A. J. Dal Canto, and S. H. Speck, J. Virol. 71:5894-5904, 1997). The predicted gene 4 product has homology to other virally encoded RCA homologs, as well as to the complement-regulatory proteins decay-accelerating factor and membrane cofactor protein. Analyses by Northern blotting and rapid amplification of cDNA ends revealed that gene 4 is transcribed as a 5.2-kb bicistronic transcript of the late kinetic class. Three gammaHV68 RCA protein isoforms (60 to 65 kDa, 50 to 55 kDa, and 40 to 45 kDa) were detected by Western blotting of infected murine NIH 3T12 fibroblast cells. A soluble 40- to 45-kDa isoform was detected in the supernatants of virally infected cells. Flow cytometric analysis revealed that the gammaHV68 RCA protein was expressed on the surfaces of infected cells. Supernatants from virally infected cells contained an activity that inhibited murine complement activation as measured by inhibition of C3 deposition on activated zymosan particles. Recombinant gammaHV68 RCA protein, containing the four conserved short consensus repeats, inhibited murine C3 deposition on zymosan via both classical and alternative pathways and inhibited deposition of human C3 on activated zymosan particles. Expression of this inhibitor of complement activation, both at the cell surface and in the fluid phase, may be important for gammaHV68 pathogenesis via the inhibition of innate and adaptive immunity.


Assuntos
Ativação do Complemento/imunologia , Gammaherpesvirinae/genética , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Linhagem Celular , Membrana Celular/metabolismo , Complemento C3/imunologia , DNA Complementar , Fibroblastos/citologia , Gammaherpesvirinae/imunologia , Expressão Gênica , Genes Virais , Humanos , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase/métodos , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Proteínas Virais/biossíntese , Proteínas Virais/imunologia
14.
Curr Opin Microbiol ; 2(4): 403-9, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10458986

RESUMO

A general association of human and primate lymphotropic herpesviruses (gamma-herpesviruses) with the development of lymphomas, as well as other tumors, especially in immunocompromised hosts, has been well documented. The lack of relevant small animal models for human gamma-herpesviruses has impeded progress in understanding the role of these viruses in the development of chronic disease. Recent research characterizing infection of inbred strains of mice with a murine gamma-herpesvirus, gamma-herpesvirus 68 (gammaHV68), is providing insights into viral and host factors involved in the establishment and control of chronic gamma-herpesvirus infection.


Assuntos
Gammaherpesvirinae/genética , Infecções por Herpesviridae/imunologia , Animais , Doença Crônica , Modelos Animais de Doenças , Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/virologia , Humanos , Camundongos , Latência Viral/genética
15.
Curr Opin Immunol ; 11(4): 371-9, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10448140

RESUMO

Murine gamma-herpesvirus 68 (gammaHV68) infection is a new model for understanding how immunity and chronic gamma-herpesvirus infection inter-relate. gammaHV68 is closely related to the human Epstein-Barr virus and Kaposi's sarcoma herpesvirus and is associated with tumors, vasculitis of the great elastic arteries and splenic fibrosis. Advances in the past year have provided an even stronger foundation for believing that gammaHV68 infection of normal and mutant mice will become the pre-eminent animal model for understanding gamma-herpesvirus pathogenesis and immunity. gammaHV68 latency has been characterized employing new assays for quantitating cells carrying the gammaHV68 genome and cells that reactivate gammaHV68 and for detecting the presence of preformed infectious virus in tissues. These advances have fostered the first steps towards a molecular definition of gammaHV68 latency. It appears that gammaHV68 shares latency programs with human gamma-herpesviruses - including the loci for gene 73, v-bcl-2 and the viral homolog of the G-protein coupled receptor. This provides candidate antigens for analysis of the role of T and B cells in regulating latency. Multiple cellular reservoirs for gammaHV68 latency were uncovered with the demonstration that gammaHV68 latently infects macrophages in addition to B cells. A critical role for B cells in regulating the nature of gammaHV68 latency was discovered and the mechanism was shown to be via alteration of the efficiency of reactivation. Studies of the response of CD4(+) and CD8(+) cells during acute and chronic gammaHV68 were performed. These new studies provide key building blocks for further development of this novel and interesting model system.


Assuntos
Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Doença Crônica , Gammaherpesvirinae/genética , Gammaherpesvirinae/fisiologia , Humanos , Interferon gama/fisiologia , Camundongos , Latência Viral
16.
J Virol ; 73(6): 4651-61, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10233924

RESUMO

The dynamics of the establishment of, and reactivation from, gammaherpesviruses latency has not been quantitatively analyzed in the natural host. Gammaherpesvirus 68 (gammaHV68) is a murine gammaherpesvirus genetically related to primate gammaherpesviruses that establishes a latent infection in infected mice. We used limiting dilution reactivation (frequency of cells reactivating gammaHV68 in vitro) and limiting dilution PCR (frequency of cells carrying gammaHV68 genome) assays to compare gammaHV68 latency in normal (C57BL/6) and B-cell-deficient (MuMT) mice. After intraperitoneal (i.p.) inoculation, latent gammaHV68 was detected in the spleen, bone marrow, and peritoneal cells. Both B-cell-deficient and C57BL/6 mice established latent infection in peritoneal cells after either i.p. or intranasal (i.n.) inoculation. In contrast, establishment of splenic latency was less efficient in B-cell-deficient than in C57BL/6 mice after i.n. inoculation. Analysis of reactivation efficiency (reactivation frequency compared to frequency of cells carrying gammaHV68 genome) revealed that (i) regardless of route or mouse strain, splenic cells reactivated gammaHV68 less efficiently than peritoneal cells, (ii) the frequency of cells carrying gammaHV68 genome was generally comparable over the course of infection between C57BL/6 and B-cell-deficient mice, (iii) between 28 and 250 days after infection, cells from B-cell-deficient mice reactivated gammaHV68 10- to 100-fold more efficiently than cells from C57BL/6 mice, (iv) at 7 weeks postinfection, B-cell-deficient mice had more genome-positive peritoneal cells than C57BL/6 mice, and (v) mixing cells (ratio of 3 to 1) that reactivated inefficiently with cells that reactivated efficiently did not significantly decrease reactivation efficiency. Consistent with a failure to normally regulate chronic gammaHV68 infection, the majority of infected B-cell-deficient mice died between 100 and 200 days postinfection. We conclude that (i) B cells are not required for establishment of gammaHV68 latency, (ii) there are organ-specific differences in the efficiency of gammaHV68 reactivation, (iii) B cells play a crucial role in regulating reactivation of gammaHV68 from latency, and (iv) B cells are important for controlling chronic gammaHV68 infection.


Assuntos
Linfócitos B/fisiologia , Gammaherpesvirinae/fisiologia , Latência Viral , Animais , Gammaherpesvirinae/genética , Genoma Viral , Infecções por Herpesviridae/virologia , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Peritônio/virologia , Especificidade da Espécie , Baço/virologia , Ativação Viral
17.
J Virol ; 73(6): 5110-22, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10233974

RESUMO

Several gammaherpesviruses contain open reading frames encoding proteins homologous to mammalian D-type cyclins. In this study, we analyzed the expression and function of the murine gammaherpesvirus 68 (gammaHV68) viral cyclin (v-cyclin). The gammaHV68 v-cyclin gene was expressed in lytically infected fibroblasts as a leaky-late mRNA of approximately 0.9 kb encoding a protein of approximately 25 kDa. To evaluate the effect of the gammaHV68 v-cyclin on cell cycle progression in primary lymphocytes and to determine if the gammaHV68 v-cyclin gene is an oncogene, we generated transgenic mice by using the lck proximal promoter to express the gammaHV68 v-cyclin in early T cells. Expression of the gammaHV68 v-cyclin significantly increased the number of thymocytes in cell culture, as determined by measuring both DNA content and incorporation of 5-bromo-2-deoxyuridine following in vivo pulse-labeling. Expression of the gammaHV68 v-cyclin interfered with normal thymocyte maturation, as shown by increased numbers of CD4(+) CD8(+) double-positive thymocytes and decreased numbers of CD4(+) or CD8(+) single-positive and T-cell-receptor-bright thymocytes and splenocytes in transgenic mice. Despite increased numbers of cycling thymocytes, gammaHV68-v-cyclin-transgenic mice did not have proportionately increased thymocyte numbers, and staining by terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling demonstrated increased apoptosis in the thymi of v-cyclin-transgenic mice. Fifteen of 38 gammaHV68-v-cyclin-transgenic mice developed high-grade lymphoblastic lymphoma between 3 and 12 months of age. We conclude that (i) the gammaHV68 v-cyclin is expressed as a leaky-late gene in lytically infected cells, (ii) expression of the gammaHV68 v-cyclin in thymocytes promotes cell cycle progression and inhibits normal T-cell differentiation, and (iii) the gammaHV68 v-cyclin gene is an oncogene.


Assuntos
Ciclinas/genética , Gammaherpesvirinae/genética , Oncogenes , Animais , Apoptose , Ciclo Celular , Ciclinas/biossíntese , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Linfoma/etiologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Coelhos , Receptores de Antígenos de Linfócitos T alfa-beta/análise , Linfócitos T/fisiologia , Proteínas Virais
18.
J Virol ; 73(5): 4524-9, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10196360

RESUMO

Several viruses, including members of the gammaherpesvirus family, encode proteins that are secreted into the extracellular environment. We have identified an abundant 44-kDa secreted protein that is present in the supernatant of fibroblasts infected with murine gammaherpesvirus 68 (gammaHV68; also referred to as MHV-68) but not in that of uninfected fibroblasts. Sequence analysis of the amino terminus and of internal peptides revealed that this protein is encoded by the gammaHV68 M3 open reading frame (ORF). The amino-terminal sequence of the secreted protein starts at residue 25 of the M3 ORF, consistent with the first 24 residues functioning as a signal peptide. Northern blot analysis revealed a single abundant approximately 1.4-kb early-late lytic transcript encoded by the M3 ORF. Analysis of a partial cDNA clone and subsequent analyses of products of rapid amplification of cDNA ends coupled with S1 nuclease protection assays demonstrate that the M3 protein is encoded by an unspliced, polyadenylated mRNA initiating at bp 7294 and terminating at bp 6007 of the gammaHV68 genome. The 3' end of the M3 transcript maps 9 bp downstream of a consensus polyadenylation signal. Thus, the predicted M3 ORF is a functional gene that encodes an abundant secreted protein which is a candidate for interacting with host cellular receptors or cytokines.


Assuntos
Gammaherpesvirinae/genética , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Viral , Camundongos , Dados de Sequência Molecular , Fases de Leitura Aberta , RNA Mensageiro , Homologia de Sequência de Aminoácidos
19.
J Virol ; 73(4): 3273-83, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10074181

RESUMO

B cells have previously been identified as the major hematopoietic cell type harboring latent gammaherpesvirus 68 (gammaHV68) (N. P. Sunil-Chandra, S. Efstathiou, and A. A. Nash, J. Gen. Virol. 73:3275-3279, 1992). However, we have shown that gammaHV68 efficiently establishes latency in B-cell-deficient mice (K. E. Weck, M. L. Barkon, L. I. Yoo, S. H. Speck, and H. W. Virgin, J. Virol. 70:6775-6780, 1996), demonstrating that B cells are not required for gammaHV68 latency. To understand this dichotomy, we determined whether hematopoietic cell types, in addition to B cells, carry latent gammaHV68. We observed a high frequency of cells that reactivate latent gammaHV68 in peritoneal exudate cells (PECs) derived from both B-cell-deficient and normal C57BL/6 mice. PECs were composed primarily of macrophages in B-cell-deficient mice and of macrophages plus B cells in normal C57BL/6 mice. To determine which cells in PECs from C57BL/6 mice carry latent gammaHV68, we developed a limiting-dilution PCR assay to quantitate the frequency of cells carrying the gammaHV68 genome in fluorescence-activated cell sorter-purified cell populations. We also quantitated the contribution of individual cell populations to the total frequency of cells carrying latent gammaHV68. At early times after infection, the frequency of PECs that reactivated gammaHV68 correlated very closely with the frequency of PECs carrying the gammaHV68 genome, validating measurement of the frequency of viral-genome-positive cells as a measure of latency in this cell population. F4/80-positive macrophage-enriched, lymphocyte-depleted PECs harbored most of the gammaHV68 genome and efficiently reactivated gammaHV68, while CD19-positive, B-cell-enriched PECs harbored about a 10-fold lower frequency of gammaHV68 genome-positive cells. CD4-positive, T-cell-enriched PECs contained only a very low frequency of gammaHV68 genome-positive cells, consistent with previous analyses indicating that T cells are not a reservoir for gammaHV68 latency (N. P. Sunil-Chandra, S. Efstathiou, and A. A. Nash, J. Gen. Virol. 73:3275-3279, 1992). Since macrophages are bone marrow derived, we determined whether elicitation of a large inflammatory response in the peritoneum would recruit additional latent cells into the peritoneum. Thioglycolate inoculation increased the total number of PECs by about 20-fold but did not affect the frequency of cells that reactivate gammaHV68, consistent with a bone marrow reservoir for latent gammaHV68. These experiments demonstrate gammaHV68 latency in two different hematopoietic cell types, F4/80-positive macrophages and CD19-positive B cells, and argue for a bone marrow reservoir for latent gammaHV68.


Assuntos
Gammaherpesvirinae/fisiologia , Macrófagos Peritoneais/virologia , Latência Viral/fisiologia , Animais , Antígenos CD , Genoma Viral , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL
20.
J Virol ; 73(3): 2321-32, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9971815

RESUMO

The program(s) of gene expression operating during murine gammaherpesvirus 68 (gammaHV68) latency is undefined, as is the relationship between gammaHV68 latency and latency of primate gammaherpesviruses. We used a nested reverse transcriptase PCR strategy (sensitive to approximately one copy of gammaHV68 genome for each genomic region tested) to screen for the presence of viral transcripts in latently infected mice. Based on the positions of known latency-associated genes in other gammaherpesviruses, we screened for the presence of transcripts corresponding to 11 open reading frames (ORFs) in the gammaHV68 genome in RNA from spleens and peritoneal cells of latently infected B-cell-deficient (MuMT) mice which have been shown contain high levels of reactivable latent gammaHV68 (K. E. Weck, M. L. Barkon, L. I. Yoo, S. H. Speck, and H. W. Virgin, J. Virol. 70:6775-6780, 1996). To control for the possible presence of viral lytic activity, we determined that RNA from latently infected peritoneal and spleen cells contained few or no detectable transcripts corresponding to seven ORFs known to encode viral gene products associated with lytic replication. However, we did detect low-level expression of transcripts arising from the region of gene 50 (encoding the putative homolog of the Epstein-Barr virus BRLF1 transactivator) in peritoneal but not spleen cells. Latently infected peritoneal cells consistently scored for expression of RNA derived from 4 of the 11 candidate latency-associated ORFs examined, including the regions of ORF M2, ORF M11 (encoding v-bcl-2), gene 73 (a homolog of the Kaposi's sarcoma-associated herpesvirus [human herpesvirus 8] gene encoding latency-associated nuclear antigen), and gene 74 (encoding a G-protein coupled receptor homolog, v-GCR). Latently infected spleen cells consistently scored positive for RNA derived from 3 of the 11 candidate latency-associated ORFs examined, including ORF M2, ORF M3, and ORF M9. To further characterize transcription of these candidate latency-associated ORFs, we examined their transcription in lytically infected fibroblasts by Northern analysis. We detected abundant transcription from regions of the genome containing ORF M3 and ORF M9, as well as the known lytic-cycle genes. However, transcription of ORF M2, ORF M11, gene 73, and gene 74 was barely detectable in lytically infected fibroblasts, consistent with a role of these viral genes during latent infection. We conclude that (i) we have identified several candidate latency genes of murine gammaHV68, (ii) expression of genes during latency may be different in different organs, consistent with multiple latency programs and/or multiple cellular sites of latency, and (iii) regions of the viral genome (v-bcl-2 gene, v-GCR gene, and gene 73) are transcribed during latency with both gammaHV68 and primate gammaherpesviruses. The implications of these findings for replacing previous operational definitions of gammaHV68 latency with a molecular definition are discussed.


Assuntos
Gammaherpesvirinae/genética , Genoma Viral , Transcrição Gênica , Latência Viral , Animais , Northern Blotting , Camundongos , Fases de Leitura Aberta , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA