Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
PLoS Pathog ; 14(2): e1006843, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29390024

RESUMO

Gammaherpesviruses encode proteins with homology to the cellular purine metabolic enzyme formyl-glycinamide-phosphoribosyl-amidotransferase (FGARAT), but the role of these viral FGARATs (vFGARATs) in the pathogenesis of a natural host has not been investigated. We report a novel role for the ORF75A vFGARAT of murine gammaherpesvirus 68 (MHV68) in infectious virion production and colonization of mice. MHV68 mutants with premature stop codons in orf75A exhibited a log reduction in acute replication in the lungs after intranasal infection, which preceded a defect in colonization of multiple host reservoirs including the mediastinal lymph nodes, peripheral blood mononuclear cells, and the spleen. Intraperitoneal infection rescued splenic latency, but not reactivation. The 75A.stop virus also exhibited defective replication in primary fibroblast and macrophage cells. Viruses produced in the absence of ORF75A were characterized by an increase in the ratio of particles to PFU. In the next round of infection this led to the alteration of early events in lytic replication including the deposition of the ORF75C tegument protein, the accelerated kinetics of viral gene expression, and induction of TNFα release and cell death. Infecting cells to deliver equivalent genomes revealed that ORF75A was required for initiating early events in infection. In contrast with the numerous phenotypes observed in the absence of ORF75A, ORF75B was dispensable for replication and pathogenesis. These studies reveal that murine rhadinovirus vFGARAT family members ORF75A and ORF75C have evolved to perform divergent functions that promote replication and colonization of the host.


Assuntos
Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/virologia , Pulmão/virologia , Macrófagos/virologia , Fases de Leitura Aberta , Baço/virologia , Proteínas Virais/metabolismo , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Códon sem Sentido , DNA Recombinante/metabolismo , DNA Viral/metabolismo , Embrião de Mamíferos/citologia , Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/patogenicidade , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Pulmão/imunologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Filogenia , Baço/imunologia , Baço/patologia , Carga Viral , Proteínas Virais/genética , Latência Viral , Replicação Viral
2.
Nucleic Acids Res ; 46(17): 9011-9026, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30137501

RESUMO

DExD/H-box helicase 9 (DHX9), or RNA helicase A (RHA), is an abundant multifunctional nuclear protein. Although it was previously reported to act as a cytosolic DNA sensor in plasmacytoid dendritic cells (pDCs), the role and molecular mechanisms of action of DHX9 in cells that are not pDCs during DNA virus infection are not clear. Here, a macrophage-specific knockout and a fibroblast-specific knockdown of DHX9 impaired antiviral innate immunity against DNA viruses, leading to increased virus replication. DHX9 enhanced NF-κB-mediated transactivation in the nucleus, which required its ATPase-dependent helicase (ATPase/helicase) domain, but not the cytosolic DNA-sensing domain. In addition, DNA virus infection did not induce cytoplasmic translocation of nuclear DHX9 in macrophages and fibroblasts. Nuclear DHX9 was associated with a multiprotein complex including both NF-κB p65 and RNA polymerase II (RNAPII) in chromatin containing NF-κB-binding sites. DHX9 was essential for the recruitment of RNAPII rather than NF-κB p65, to the corresponding promoters; this function also required its ATPase/helicase activity. Taken together, our results show a critical role of nuclear DHX9 (as a transcription coactivator) in the stimulation of NF-κB-mediated innate immunity against DNA virus infection, independently of DHX9's DNA-sensing function.


Assuntos
RNA Helicases DEAD-box/genética , DNA Viral/genética , Interações Hospedeiro-Patógeno/genética , Imunidade Inata , NF-kappa B/genética , RNA Polimerase II/genética , Animais , Chlorocebus aethiops , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/imunologia , DNA Viral/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Feminino , Gammaherpesvirinae/genética , Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/imunologia , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/imunologia , Células-Tronco Embrionárias Murinas/virologia , NF-kappa B/imunologia , Células NIH 3T3 , Cultura Primária de Células , RNA Polimerase II/imunologia , Transdução de Sinais , Células Vero , Replicação Viral
3.
J Biol Chem ; 293(8): 2801-2814, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29305424

RESUMO

The stress-induced unfolded protein response (UPR) in the endoplasmic reticulum (ER) involves various signaling cross-talks and controls cell fate. B-cell receptor (BCR) signaling, which can trigger UPR, induces gammaherpesvirus lytic replication and serves as a physiological mechanism for gammaherpesvirus reactivation in vivo However, how the UPR regulates BCR-mediated gammaherpesvirus infection is unknown. Here, we demonstrate that the ER stressors tunicamycin and thapsigargin inhibit BCR-mediated murine gammaherpesvirus 68 (MHV68) lytic replication by inducing expression of the UPR mediator Bip and blocking activation of Akt, ERK, and JNK. Both Bip and the downstream transcription factor ATF4 inhibited BCR-mediated MHV68 lytic gene expression, whereas UPR-induced C/EBP homologous protein (CHOP) was required for and promoted BCR-mediated MHV68 lytic replication by suppressing upstream Bip and ATF4 expression. Bip knockout was sufficient to rescue BCR-mediated MHV68 lytic gene expression in CHOP knockout cells, and this rescue was blocked by ectopic ATF4 expression. Furthermore, ATF4 directly inhibited promoter activity of the MHV68 lytic switch transactivator RTA. Altogether, we show that ER stress-induced CHOP inhibits Bip and ATF4 expression and that ATF4, in turn, plays a critical role in CHOP-mediated regulation of BCR-controlled MHV68 lytic replication. We conclude that ER stress-mediated UPR and BCR signaling pathways are interconnected and form a complex network to regulate the gammaherpesvirus infection cycle.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Linfócitos B/virologia , Estresse do Retículo Endoplasmático , Gammaherpesvirinae/fisiologia , Proteínas de Choque Térmico/metabolismo , Receptores de Antígenos de Linfócitos B/agonistas , Fator de Transcrição CHOP/metabolismo , Fator 4 Ativador da Transcrição/antagonistas & inibidores , Fator 4 Ativador da Transcrição/genética , Animais , Antivirais/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linhagem Celular Transformada , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Gammaherpesvirinae/efeitos dos fármacos , Gammaherpesvirinae/crescimento & desenvolvimento , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/genética , Lisogenia/efeitos dos fármacos , Camundongos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia , Fator de Transcrição CHOP/antagonistas & inibidores , Fator de Transcrição CHOP/genética , Tunicamicina/farmacologia , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ativação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
4.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795415

RESUMO

Gammaherpesviruses are ubiquitous pathogens that establish lifelong infection in >95% of adults worldwide and are associated with a variety of malignancies. Coevolution of gammaherpesviruses with their hosts has resulted in an intricate relationship between the virus and the host immune system, and perturbation of the virus-host balance results in pathology. Interferon regulatory factor 1 (IRF-1) is a tumor suppressor that is also involved in the regulation of innate and adaptive immune responses. Here, we show that type I interferon (IFN) and IRF-1 cooperate to control acute gammaherpesvirus infection. Specifically, we demonstrate that a combination of IRF-1 and type I IFN signaling ensures host survival during acute gammaherpesvirus infection and supports IFN gamma-mediated suppression of viral replication. Thus, our studies reveal an intriguing cross talk between IRF-1 and type I and II IFNs in the induction of the antiviral state during acute gammaherpesvirus infection. IMPORTANCE: Gammaherpesviruses establish chronic infection in a majority of adults, and this long-term infection is associated with virus-driven development of a range of malignancies. In contrast, a brief period of active gammaherpesvirus replication during acute infection of a naive host is subclinical in most individuals. Here, we discovered that a combination of type I interferon (IFN) signaling and interferon regulatory factor 1 (IRF-1) expression is required to ensure survival of a gammaherpesvirus-infected host past the first 8 days of infection. Specifically, both type I IFN receptor and IRF-1 expression potentiated antiviral effects of type II IFN to restrict gammaherpesvirus replication in vivo, in the lungs, and in vitro, in primary macrophage cultures.


Assuntos
Gammaherpesvirinae/patogenicidade , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno , Fator Regulador 1 de Interferon/genética , Interferon-alfa/genética , Interferon beta/genética , Animais , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/imunologia , Antígenos Nucleares/genética , Antígenos Nucleares/imunologia , Gammaherpesvirinae/crescimento & desenvolvimento , Regulação da Expressão Gênica , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/mortalidade , Infecções por Herpesviridae/virologia , Humanos , Fator Regulador 1 de Interferon/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Interferon gama/genética , Interferon gama/imunologia , Pulmão/imunologia , Pulmão/virologia , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cultura Primária de Células , Proteínas/genética , Proteínas/imunologia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais , Baço/imunologia , Baço/virologia , Análise de Sobrevida
5.
J Exp Med ; 195(7): 931-40, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11927636

RESUMO

Many gamma-herpesviruses encode candidate oncogenes including homologues of host bcl-2 and cyclin proteins (v-bcl-2, v-cyclin), but the physiologic roles of these genes during infection are not known. We show for the first time in any virus system the physiologic role of v-bcl-2. A gamma-herpesvirus v-bcl-2 was essential for efficient ex vivo reactivation from latent infection, and for both persistent replication and virulence during chronic infection of immunocompromised (interferon [IFN]-gamma(-/-)) mice. The v-cyclin was also critical for the same stages in pathogenesis. Strikingly, while the v-bcl-2 and v-cyclin were important for chronic infection, these genes were not essential for viral replication in cell culture, viral replication during acute infection in vivo, establishment of latent infection, or virulence during acute infection. We conclude that v-bcl-2 and v-cyclin have important roles during latent and persistent gamma-herpesvirus infection and that herpesviruses encode genes with specific roles during chronic infection and disease, but not acute infection and disease. As gamma-herpesviruses primarily cause human disease during chronic infection, these chronic disease genes may be important targets for therapeutic intervention.


Assuntos
Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/genética , Genes bcl-2 , Infecções por Herpesviridae/genética , Animais , Linhagem Celular , Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Proteínas Oncogênicas/genética , Reação em Cadeia da Polimerase , Fatores de Tempo , Ativação Viral , Latência Viral
6.
J Gen Virol ; 91(Pt 4): 867-79, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19940063

RESUMO

Two novel gammaherpesviruses were isolated, one from a field vole (Microtus agrestis) and the other from wood mice (Apodemus sylvaticus). The genome of the latter, designated wood mouse herpesvirus (WMHV), was completely sequenced. WMHV had the same genome structure and predicted gene content as murid herpesvirus 4 (MuHV4; murine gammaherpesvirus 68). Overall nucleotide sequence identity between WMHV and MuHV4 was 85 % and most of the 10 kb region at the left end of the unique region was particularly highly conserved, especially the viral tRNA-like sequences and the coding regions of genes M1 and M4. The partial sequence (71 913 bp) of another gammaherpesvirus, Brest herpesvirus (BRHV), which was isolated ostensibly from a white-toothed shrew (Crocidura russula), was also determined. The BRHV sequence was 99.2 % identical to the corresponding portion of the WMHV genome. Thus, WMHV and BRHV appeared to be strains of a new virus species. Biological characterization of WMHV indicated that it grew with similar kinetics to MuHV4 in cell culture. The pathogenesis of WMHV in wood mice was also extremely similar to that of MuHV4, except for the absence of inducible bronchus-associated lymphoid tissue at day 14 post-infection and a higher load of latently infected cells at 21 days post-infection.


Assuntos
Arvicolinae/virologia , Gammaherpesvirinae/classificação , Murinae/virologia , Rhadinovirus/classificação , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Viral/química , Gammaherpesvirinae/genética , Gammaherpesvirinae/crescimento & desenvolvimento , Genoma Viral , Dados de Sequência Molecular , Rhadinovirus/genética , Rhadinovirus/crescimento & desenvolvimento , Proteínas da Matriz Viral/análise , Proteínas da Matriz Viral/genética
7.
J Virol ; 83(20): 10582-95, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656880

RESUMO

Tegument is a unique structure of herpesvirus, which surrounds the capsid and interacts with the envelope. Morphogenesis of gammaherpesvirus is poorly understood due to lack of efficient lytic replication for Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus/human herpesvirus 8, which are etiologically associated with several types of human malignancies. Murine gammaherpesvirus 68 (MHV-68) is genetically related to the human gammaherpesviruses and presents an excellent model for studying de novo lytic replication of gammaherpesviruses. MHV-68 open reading frame 33 (ORF33) is conserved among Alpha-, Beta-, and Gammaherpesvirinae subfamilies. However, the specific role of ORF33 in gammaherpesvirus replication has not yet been characterized. We describe here that ORF33 is a true late gene and encodes a tegument protein. By constructing an ORF33-null MHV-68 mutant, we demonstrated that ORF33 is not required for viral DNA replication, early and late gene expression, viral DNA packaging or capsid assembly but is required for virion morphogenesis and egress. Although the ORF33-null virus was deficient in release of infectious virions, partially tegumented capsids produced by the ORF33-null mutant accumulated in the cytoplasm, containing conserved capsid proteins, ORF52 tegument protein, but virtually no ORF45 tegument protein and the 65-kDa glycoprotein B. Finally, we found that the defect of ORF33-null MHV-68 could be rescued by providing ORF33 in trans or in an ORF33-null revertant virus. Taken together, our results indicate that ORF33 is a tegument protein required for viral lytic replication and functions in virion morphogenesis and egress.


Assuntos
Gammaherpesvirinae/crescimento & desenvolvimento , Fases de Leitura Aberta/fisiologia , Vírion , Animais , Linhagem Celular , Citoplasma/metabolismo , Citoplasma/ultraestrutura , Gammaherpesvirinae/genética , Gammaherpesvirinae/metabolismo , Gammaherpesvirinae/ultraestrutura , Humanos , Microscopia Eletrônica de Transmissão , Morfogênese , Mutação , Fases de Leitura Aberta/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Vírion/ultraestrutura , Montagem de Vírus
8.
Tuberculosis (Edinb) ; 116: 56-60, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31153519

RESUMO

Tuberculosis is caused by Mycobacterium tuberculosis (Mtb), a bacterial pathogen which is transmitted via aerosol and establishes a chronic lung infection. In naïve hosts, Mtb grows for several weeks without being restricted by IFNγ-producing T cells, which eventually accumulate and limit Mtb dissemination. In this study, we used a mouse model of Mtb/γ-herpesvirus (γHV) coinfection to test the hypothesis that latent γHV infection alters host resistance to Mtb. γHVs are DNA viruses which elicit a polyclonal T cell response and attenuate some acute bacterial pathogens in mice; whether γHVs modulate infection with Mtb is unknown. Here, mice harboring latent mouse gammaherpesvirus 68 (MHV68)-a γHV genetically and biologically related to human Epstein Barr virus (EBV)-were infected via aerosol with a low dose of virulent Mtb. Mtb burdens and IFNγ+ T cell frequencies in mice with latent MHV68 (MHV68POS mice) were subsequently measured and compared to control mice that did not harbor latent MHV68 (MHV68NEG mice). Relative to MHV68NEG controls, MHV68POS mice more effectively limited Mtb growth and dissemination, and had higher frequencies of CD4+IFNγ+ cells in lung-draining lymph nodes. Collectively, our results support a model wherein latent γHV confers moderate protection against subsequent Mtb infection.


Assuntos
Coinfecção , Gammaherpesvirinae/patogenicidade , Infecções por Herpesviridae/virologia , Mycobacterium tuberculosis/patogenicidade , Tuberculose/microbiologia , Latência Viral , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/virologia , Modelos Animais de Doenças , Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno , Interferon gama/imunologia , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/imunologia , Fatores de Tempo , Tuberculose/imunologia , Tuberculose/prevenção & controle
9.
Brain Behav Immun ; 22(6): 912-22, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18280699

RESUMO

While Ecstasy (3,4-methylenedioxymethamphetamine, MDMA) has been shown to modulate immune responses, no studies have addressed drug-induced alterations to viral infection. In this study, bone marrow-derived macrophages were exposed to MDMA, then infected with murine gammaherpesvirus-68, and the expression of monokines assessed. MDMA-induced reductions in virus-stimulated monokine mRNA expression were observed in a dose-dependent manner. In particular, IL-6 mRNA expression and secretion was significantly decreased in gammaherpesvirus-infected macrophages exposed to MDMA. Concentrations of MDMA capable of reducing monokine production did not induce significant cell death and allowed normal viral gene expression. These studies represent the first to demonstrate the ability of this drug of abuse to alter a viral-induced macrophage response.


Assuntos
Gammaherpesvirinae/crescimento & desenvolvimento , Macrófagos/efeitos dos fármacos , Monocinas/genética , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/biossíntese , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Monocinas/biossíntese , Monocinas/metabolismo , N-Metil-3,4-Metilenodioxianfetamina/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Acta Virol ; 43(2-3): 186-91, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10696443

RESUMO

Despite reliance on the need to continually prepare fresh cultures of chick embryo fibroblasts (CEFs) to make Marek's disease (MD) vaccines, MD vaccines are the most widely used vaccines in the poultry industry. Preparation of CEF's accounts for approximately 40% of the costs associated with producing MD vaccines. A significant reduction in MD vaccine production costs could be realized if a continuous cell lines were available for MD vaccine production. Recently, we reported development and characterization of a cell line system (OCL) that supports growth and replication of oncogenic serotype 1 Marek's disease virus (MDV). Here we report development of three cell line systems for production of MD vaccine. These cell lines support the growth and replication of attenuated serotype 1 MDV (CVI-OCL), serotype 2 MDV (SB1-OCL) and serotype 3 MDV (HVT-OCL). MDV is maintained in a stable state in the OCL cells and the infected cells can be continuously grown. The vaccines made from these cell lines are safe and protect White Leghorn chickens against challenge with very virulent serotype 1 MDV, similar to traditional vaccines made from CEF cells. These cell line systems can significantly reduce the costs associated with MD vaccine production. Furthermore, the increased stability of MDV and the potential for positive selection of recombinant MDV suggest that OCL will be ideal for production of more effective MDV vaccines using recombinant DNA technology.


Assuntos
Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/imunologia , Herpesvirus Galináceo 2/crescimento & desenvolvimento , Herpesvirus Galináceo 2/imunologia , Vacinas Virais/imunologia , Cultura de Vírus , Animais , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Galinhas , Fibroblastos , Doença de Marek/prevenção & controle , Perus/virologia , Ensaio de Placa Viral , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos
11.
Bull Mem Acad R Med Belg ; 154(6 Pt 2): 363-9, 1999.
Artigo em Francês | MEDLINE | ID: mdl-10992885

RESUMO

This work was devoted to the study of the interactions between bovine herpesvirus 4 (BHV-4) and bovine cells in vitro. It led to the discovery of two interesting properties of BVH-4 replication cycle: first, the cellular receptor heparan sulfate was proven to mediate BVH-4 binding to target cells. This is the first description of the implication of heparan sulfate in the binding process of a gammaherpesvirus. Second, using synchronised cells, the replication of BVH-4 DNA was proven to be dependent on the S phase of the cell cycle. This dependence could explain some properties of BVH-4 infection in vitro and could play an important role in the biology of the infection in vivo. Finally, in order to produce monoclonal antibodies against BVH-4 IE1 and IE2 proteins, the genes coding for these proteins were cloned and expressed in prokaryotic cells.


Assuntos
Doenças dos Bovinos/virologia , Gammaherpesvirinae/crescimento & desenvolvimento , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Integração Viral/fisiologia , Replicação Viral/fisiologia , Animais , Sítios de Ligação , Bovinos , DNA Viral/genética , DNA Viral/fisiologia , Gammaherpesvirinae/genética , Gammaherpesvirinae/imunologia , Heparitina Sulfato/fisiologia , Fase S/fisiologia , Cultura de Vírus
12.
Viral Immunol ; 23(6): 547-55, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21142440

RESUMO

Infection of mice with murine gammaherpesvirus-68 (γHV-68) serves as a model to understand the pathogenesis of persistent viral infections, including the potential for co-infections to modulate viral latency. We have previously found that infection of neonates (8-day-old mice) with γHV-68 resulted in a high level of persistence of the virus in the lungs as well as the spleen, in contrast to infection of adult mice, for which long-term latency was only readily detected in the spleen. In this study we investigated whether stimulation of toll-like receptor (TLR)9 would modulate viral latency in mice infected with γHV-68 in an age-dependent manner. Pups and adult mice were injected with the synthetic TLR9 ligand CpG ODN at 30 dpi, at which time long-term latency has been established. Three days after CpG injection, the lungs and spleens were removed, and a limiting dilution assay was done to determine the frequency of latently infected cells. RNA was extracted to measure viral transcripts using a ribonuclease protection assay. We observed that CpG injection resulted in an increase in the frequency of latently-infected cells in both the lungs and spleens of infected pups, but only in the spleens of infected adult mice. No preformed virus was detected, suggesting that TLR9 stimulation did not trigger complete viral reactivation. When we examined viral gene expression in these same tissues, we observed expression only of the immediate early lytic genes, rta and K3, but not the early DNA polymerase gene or late gB transcript indicative of an abortive reactivation in the spleen. Additionally, mice infected as pups had greater numbers of germinal center B cells in the spleen following CpG injection, whereas CpG stimulated the expansion of follicular zone B cells in adult mice. These data suggest that stimulation of TLR9 differentially modulates gammaherpesvirus latency via an age-dependent mechanism.


Assuntos
Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/virologia , Receptor Toll-Like 9/metabolismo , Ativação Viral , Latência Viral , Envelhecimento/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Gammaherpesvirinae/genética , Gammaherpesvirinae/crescimento & desenvolvimento , Regulação Viral da Expressão Gênica , Pulmão/virologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/administração & dosagem , Baço/virologia , Receptor Toll-Like 9/imunologia
13.
PLoS One ; 5(7): e11672, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20657771

RESUMO

BACKGROUND: Viruses have evolved to evade the host's complement system. The open reading frames 4 (ORF4) of gammaherpesviruses encode homologs of regulators of complement activation (RCA) proteins, which inhibit complement activation at the level of C3 and C4 deposition. Besides complement regulation, these proteins are involved in heparan sulfate and glycosaminoglycan binding, and in case of MHV-68, also in viral DNA synthesis in macrophages. METHODOLOGY/PRINCIPAL FINDINGS: Here, we made use of MHV-68 to study the role of ORF4 during infection of fibroblasts. While attachment and penetration of virions lacking the RCA protein were not affected, we observed a delayed delivery of the viral genome to the nucleus of infected cells. Analysis of the phosphorylation status of a variety of kinases revealed a significant reduction in phosphorylation of the protein kinase Akt in cells infected with ORF4 mutant virus, when compared to cells infected with wt virus. Consistent with a role of Akt activation in initial stages of infection, inhibition of Akt signaling in wt virus infected cells resulted in a phenotype resembling the phenotype of the ORF4 mutant virus, and activation of Akt by addition of insulin partially reversed the phenotype of the ORF4 mutant virus. Importantly, the homologous ORF4 of KSHV was able to rescue the phenotype of the MHV-68 ORF4 mutant, indicating that ORF4 is functionally conserved and that ORF4 of KSHV might have a similar function in infection initiation. CONCLUSIONS/SIGNIFICANCE: In summary, our studies demonstrate that ORF4 contributes to efficient infection by activation of the protein kinase Akt and thus reveal a novel function of a gammaherpesvirus RCA protein.


Assuntos
Gammaherpesvirinae/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Virais/fisiologia , Animais , Citometria de Fluxo , Gammaherpesvirinae/genética , Gammaherpesvirinae/crescimento & desenvolvimento , Teste de Complementação Genética , Insulina/farmacologia , Cinética , Camundongos , Microscopia de Fluorescência , Células NIH 3T3 , Fases de Leitura Aberta/genética , Fosforilação/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleosídeos/farmacologia , Transdução de Sinais , Proteínas Virais/genética
14.
PLoS One ; 3(3): e1673, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-18320026

RESUMO

BACKGROUND: The tachykinins are implicated in neurogenic inflammation and the neuropeptide substance P in particular has been shown to be a proinflammatory mediator. A role for the tachykinins in host response to lung challenge has been previously demonstrated but has been focused predominantly on the release of the tachykinins from nerves innervating the lung. We have previously demonstrated the most dramatic phenotype described for the substance P encoding gene preprotachykinin-A (PPT-A) to date in controlling the host immune response to the murine gammaherpesvirus 68, in the lung. METHODOLOGY/PRINCIPAL FINDINGS: In this study we have utilised transgenic mice engineered to co-ordinately express the beta-galactosidase marker gene along with PPT-A to facilitate the tracking of PPT-A expression. Using a combination of these mice and conventional immunohistology we now demonstrate that PPT-A gene expression and substance P peptide are induced in cells of the respiratory tract including tracheal, bronchiolar and alveolar epithelial cells and macrophages after viral infection. This induction was observed 24h post infection, prior to observable inflammation and the expression of pro-inflammatory chemokines in this model. Induced expression of the PPT-A gene and peptide persisted in the lower respiratory tract through day 7 post infection. CONCLUSIONS/SIGNIFICANCE: Non-neuronal PPT-A expression early after infection may have important clinical implications for the progression or management of lung disease or infection aside from the well characterised later involvement of the tachykinins during the inflammatory response.


Assuntos
Epitélio/metabolismo , Gammaherpesvirinae/genética , Infecções por Herpesviridae/metabolismo , Precursores de Proteínas/genética , Sistema Respiratório/metabolismo , Taquicininas/metabolismo , Animais , Epitélio/virologia , Gammaherpesvirinae/crescimento & desenvolvimento , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Técnicas Imunoenzimáticas , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Precursores de Proteínas/metabolismo , Sistema Respiratório/citologia , Sistema Respiratório/virologia , Substância P/metabolismo , Taquicininas/genética , beta-Galactosidase/metabolismo
15.
Vaccine ; 25(20): 3934-45, 2007 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-17433507

RESUMO

Efficient vaccines against AIDS, Hepatitis C and other persistent virus infections are urgently needed. Vaccine development has been especially hampered by the lack of suitable small animal models to reliably test the protective capacity of candidate vaccines against such chronic viral infections. A natural mouse pathogen such as MHV-68 that persists lifelong after infection, appears to be a particularly promising candidate for a more relevant model system. Here, we investigated infections with recombinant MHV-68 as novel mouse challenge model to test the efficacy of heterologous vaccines based on recombinant modified vaccinia virus Ankara (MVA). To apply ovalbumin (OVA) as a model antigen, we constructed the recombinant virus MHV-68-OVA by BAC technology and characterized genetic stability and replicative capacity of the virus in vitro and in vivo. We demonstrated the ability of MHV-68-OVA to produce ovalbumin upon tissue culture infection. Moreover, the use of MHV-68-OVA-infected target cells allowed for efficient ex vivo amplification of OVA-specific, MHC class I-restricted CD8 T cells derived from MVA-OVA-vaccinated C57BL/6 mice. Finally, we immunized C57BL/6 mice with MVA-OVA and challenged the animals with MHV-68-OVA testing different time points and routes of infection. Vaccinated mice were infected with MHV-68-OVA but showed reduced viral loads in the acute and latent phase of challenge infection. These data strongly suggest the usefulness of the MHV-68 challenge model for further evaluation of recombinant vaccines against persisting virus infections.


Assuntos
Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/prevenção & controle , Vacinas contra Herpesvirus/farmacologia , Ovalbumina/imunologia , Vaccinia virus/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Galinhas , Modelos Animais de Doenças , Gammaherpesvirinae/genética , Gammaherpesvirinae/crescimento & desenvolvimento , Genes MHC Classe I/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Vacinas contra Herpesvirus/genética , Vacinas contra Herpesvirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/farmacologia , Vaccinia virus/genética , Carga Viral
16.
Virus Genes ; 32(1): 27-35, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16525732

RESUMO

Ovine herpesvirus 2 (OvHV-2) is a lymphotropic gammaherpesvirus that asymptomatically infects most sheep, but causes malignant catarrhal fever in cattle, bison, pigs and deer. There is no permissive cell culture system but OvHV-2-infected T lymphocytes can be cultured from diseased animals. We showed that the OvHV-2 genome was in a circular conformation in sheep peripheral blood mononuclear cells and that the latency-associated ORF73 was transcribed, while expression of the productive cycle genes ORF9 (DNA polymerase) and ORF50 (R-transactivator) was barely detectable, suggestive of latency. Doxorubicin treatment of these cells induced the appearance of linear viral DNA and transcription of productive cycle genes along with several viral unique genes. In contrast, cultured T cells from diseased cattle and rabbits contained a mixture of circular and linear genome configurations indicative of a mixture of latently- and productively-infected cells. Most of the OvHV-2 unique genes were transcribed in these cells but ORF50 expression was only seen after doxorubicin treatment indicating a 'leaky' latent pattern of gene expression. 5-azacytidine treatment increased the proportion of circular DNA and inhibited the expression of most of the OvHV-2 unique genes except Ov2.5 (vIL-10) and Ov4.5 (Bcl-2 homologue) in the cattle cell line. These studies provide key insights into the differences in OvHV-2 gene expression in cells from reservoir and susceptible species and, for the first time, an in vitro system for studying the latent and productive phases of the OvHV-2 virus life cycle.


Assuntos
Gammaherpesvirinae/genética , Genes Virais , Animais , Azacitidina/farmacologia , Sequência de Bases , Bovinos , Linhagem Celular , DNA Viral/química , DNA Viral/genética , Reservatórios de Doenças , Doxorrubicina/farmacologia , Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/patogenicidade , Expressão Gênica/efeitos dos fármacos , Conformação de Ácido Nucleico , Coelhos , Ovinos , Especificidade da Espécie , Linfócitos T/virologia , Transcrição Gênica
17.
J Gen Virol ; 80 ( Pt 10): 2729-2736, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10573167

RESUMO

Primary infection with murine gammaherpesvirus-68 (MHV-68), as with other members of the gammaherpesvirus subfamily, is characterized by a lymphoproliferative phase. MHV-68 causes acute splenomegaly and an infectious mononucleosis-like syndrome in which there is expansion of the CD8+ T cell subset. In long-term infections, MHV-68 is associated with lymphoma development. In order to elucidate the mechanisms underlying the proliferative processes, the events following infection of murine splenocytes or purified murine B lymphocytes in vitro have been examined. MHV-68 infection prolonged the viability of murine splenocytes and stimulated cellular proliferation. Unlike Epstein-Barr virus and herpesvirus saimiri, MHV-68 did not cause growth transformation. Growth transformation did not occur even when cells with a predisposition to transformation were infected or when culture conditions were selected to enhance the viability of the cells. Following MHV-68 infection, the latency-associated viral tRNAs were transcribed. However, transcription of the other known latency-associated gene, M2, was not observed. In addition, there was no evidence of productive virus replication either by staining with antibodies specific for late virus antigens or by in situ hybridization for early and late mRNAs. In contrast to Epstein-Barr virus- and herpesvirus saimiri-infected lymphocytes, where episomal genomes are seen, Gardella gel analysis indicated that the primary lymphocytes infected by MHV-68 in vitro contained only linear virus DNA. This DNA was nuclease sensitive, indicating that, while MHV-68 was efficiently uncoated, its circularization in vitro was extremely inefficient. These results are discussed in terms of the host-virus interaction.


Assuntos
Linfócitos B/virologia , Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/genética , Animais , Linhagem Celular , Transformação Celular Viral , Células Cultivadas , Cricetinae , Feminino , Genoma Viral , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Conformação de Ácido Nucleico , Fenótipo , Transcrição Gênica
18.
J Gen Virol ; 85(Pt 1): 137-146, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14718628

RESUMO

A regulated switch between latent and lytic gene expression is common to all known herpesviruses. However, the effects on host colonization of altering this switch are largely unknown. We deregulated the transcription of the gene encoding the major lytic transactivator of murine gammaherpesvirus-68, ORF50, by inserting a new and powerful promoter element in its 5' untranslated region. In vitro, the mutant virus (M50) transcribed ORF50 at a high level and showed more rapid lytic spread in permissive fibroblast cultures, but in vivo, the M50 virus showed a severe deficit in latency establishment, with no sign of the infectious mononucleosis-like illness normally associated with wild-type infection. Although a low level of M50 viral DNA was detectable by PCR in spleens, replication-competent virus could not be recovered beyond 10 days post-infection. The M50 virus was also attenuated in immunocompromised mice. Thus a gammaherpesvirus unable to shut off lytic cycle gene expression showed severely restricted host colonization.


Assuntos
Gammaherpesvirinae/crescimento & desenvolvimento , Gammaherpesvirinae/fisiologia , Gammaherpesvirinae/patogenicidade , Regulação Viral da Expressão Gênica , Mutação , Latência Viral , Replicação Viral , Animais , Sequência de Bases , Linhagem Celular , Cricetinae , Feminino , Gammaherpesvirinae/genética , Infecções por Herpesviridae/virologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Células NIH 3T3 , Fases de Leitura Aberta/genética , Proteínas Virais/genética , Virulência , Ativação Viral , Latência Viral/genética
19.
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
20.
Proc Natl Acad Sci U S A ; 98(11): 6325-9, 2001 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-11353832

RESUMO

Reactivation of latent herpesviruses is a particular problem in immunocompromised individuals, such as AIDS patients, who lack effective CD4 T helper cell function. An important question is whether residual immune defenses can be mobilized to combat such opportunistic infections, in the absence of CD4 T cells. In the present study, we used a mouse model of opportunistic infection to determine whether stimulation via CD40 could substitute for CD4 T cell function in preventing reactivation of a latent herpesvirus. Treatment with an agonistic antibody to CD40 was highly effective in preventing reactivation of latent murine gammaherpesvirus (MHV-68) in the lungs of CD4 T cell-deficient mice. CD8(+) T cells were essential for this effect, whereas virus-specific serum antibody was undetectable and IFN-gamma production was unchanged. This demonstration that immunostimulation via CD40 can replace CD4 T cell help in controlling latent virus in vivo has potential implications for the development of novel therapeutic agents to prevent viral reactivation in immunocompromised patients.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Gammaherpesvirinae/imunologia , Ativação Viral/imunologia , Latência Viral/imunologia , Células 3T3 , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Gammaherpesvirinae/crescimento & desenvolvimento , Interferon gama/biossíntese , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA