Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 12(11)2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33126536

RESUMO

Cytomegaloviruses all encode the viral inhibitor of caspase-8-induced apoptosis (vICA). After binding to this initiator caspase, vICA blocks caspase-8 proteolytic activity and ability to activate caspase-3 and/or caspase-7. In this manner, vICA has long been known to prevent apoptosis triggered via tumor necrosis factor (TNF) family death receptor-dependent extrinsic signaling. Here, we employ fully wild-type murine cytomegalovirus (MCMV) and vICA-deficient MCMV (∆M36) to investigate the contribution of TNF signaling to apoptosis during infection of different cell types. ∆M36 shows the expected ability to kill mouse splenic hematopoietic cells, bone marrow-derived macrophages (BMDM), and dendritic cells (BMDC). Antibody blockade or genetic elimination of TNF protects myeloid cells from death, and caspase-8 activation accompanies cell death. Interferons, necroptosis, and pyroptotic gasdermin D (GSDMD) do not contribute to myeloid cell death. Human and murine fibroblasts or murine endothelial cells (SVEC4-10) normally insensitive to TNF become sensitized to ∆M36-induced apoptosis when treated with TNF or TNF-containing BMDM-conditioned medium. We demonstrate that myeloid cells are the natural source of TNF that triggers apoptosis in either myeloid (autocrine) or non-myeloid cells (paracrine) during ∆M36 infection of mice. Caspase-8 suppression by vICA emerges as key to subverting innate immune elimination of a wide variety of infected cell types.


Assuntos
Apoptose/genética , Caspase 8/metabolismo , Muromegalovirus/patogenicidade , Transdução de Sinais , Fator de Necrose Tumoral alfa/imunologia , Proteínas Virais/genética , Animais , Caspase 8/genética , Sobrevivência Celular , Células Dendríticas/imunologia , Células Dendríticas/virologia , Células Endoteliais/imunologia , Células Endoteliais/virologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Knockout , Muromegalovirus/imunologia , Células Mieloides/imunologia , Células Mieloides/virologia , Células NIH 3T3 , Fator de Necrose Tumoral alfa/genética
2.
Proc Natl Acad Sci U S A ; 114(13): E2786-E2795, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28292903

RESUMO

The complex interplay between caspase-8 and receptor-interacting protein (RIP) kinase RIP 3 (RIPK3) driving extrinsic apoptosis and necroptosis is not fully understood. Murine cytomegalovirus triggers both apoptosis and necroptosis in infected cells; however, encoded inhibitors of caspase-8 activity (M36) and RIP3 signaling (M45) suppress these antiviral responses. Here, we report that this virus activates caspase-8 in macrophages to trigger apoptosis that gives rise to secondary necroptosis. Infection with double-mutant ΔM36/M45mutRHIM virus reveals a signaling pattern in which caspase-8 activates caspase-3 to drive apoptosis with subsequent RIP3-dependent activation of mixed lineage kinase domain-like (MLKL) leading to necroptosis. This combined cell death signaling is highly inflammatory, greater than either apoptosis induced by ΔM36 or necroptosis induced by M45mutRHIM virus. IL-6 production by macrophages is dramatically increased during double-mutant virus infection and correlates with faster antiviral responses in the host. Collaboratively, M36 and M45 target caspase-8 and RIP3 pathways together to suppress this proinflammatory cell death. This study reveals the effect of antiviral programmed cell death pathways on inflammation, shows that caspase-8 activation may go hand-in-hand with necroptosis in macrophages, and revises current understanding of independent and collaborative functions of M36 and M45 in blocking apoptotic and necroptotic cell death responses.


Assuntos
Apoptose , Infecções por Herpesviridae/veterinária , Muromegalovirus/metabolismo , Ribonucleotídeo Redutases/metabolismo , Doenças dos Roedores/fisiopatologia , Proteínas Virais/metabolismo , Animais , Caspase 8/genética , Caspase 8/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/fisiopatologia , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno , Camundongos , Muromegalovirus/classificação , Muromegalovirus/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Ribonucleotídeo Redutases/genética , Doenças dos Roedores/genética , Doenças dos Roedores/imunologia , Doenças dos Roedores/virologia , Proteínas Virais/genética
3.
Cell Mol Immunol ; 12(2): 170-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25544503

RESUMO

A universal cytomegalovirus (CMV) vaccination promises to reduce the burden of the developmental damage that afflicts up to 0.5% of live births worldwide. An effective vaccination that prevents transplacental transmission would reduce CMV congenital disease and CMV-associated still births and leave populations less susceptible to opportunistic CMV disease. Thus, a vaccination against this virus has long been recognized for the potential of enormous health-care savings because congenital damage is life-long and existing anti-viral options are limited. Vaccine researchers, industry leaders, and regulatory representatives have discussed the challenges posed by clinical efficacy trials that would lead to a universal CMV vaccine, reviewing the links between infection and disease, and identifying settings where disrupting viral transmission might provide a surrogate endpoint for disease prevention. Reducing the complexity of such trials would facilitate vaccine development. Children and adolescents are the targets for universal vaccination, with the expectation of protecting the offspring of immunized women. Given that a majority of females worldwide experience CMV infection during childhood, a universal vaccine must boost natural immunity and reduce transmission due to reactivation and re-infection as well as primary infection during pregnancy. Although current vaccine strategies recognize the value of humoral and cellular immunity, the precise mechanisms that act at the placental interface remain elusive. Immunity resulting from natural infection appears to limit rather than prevent reactivation of latent viruses and susceptibility to re-infection, leaving a challenge for universal vaccination to improve upon natural immunity levels. Despite these hurdles, early phase clinical trials have achieved primary end points in CMV seronegative subjects. Efficacy studies must be expanded to mixed populations of CMV-naive and naturally infected subjects to understand the overall efficacy and potential. Together with CMV vaccine candidates currently in clinical development, additional promising preclinical strategies continue to come forward; however, these face limitations due to the insufficient understanding of host defense mechanisms that prevent transmission, as well as the age-old challenges of reaching the appropriate threshold of immunogenicity, efficacy, durability and potency. This review focuses on the current understanding of natural and CMV vaccine-induced protective immunity.


Assuntos
Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/uso terapêutico , Citomegalovirus/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Humanos , Vacinação
4.
J Virol ; 87(20): 11107-20, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23926341

RESUMO

Human cytomegalovirus (HCMV), a betaherpesvirus, can cause severe disease in immunosuppressed patients and following congenital infection. A vaccine that induces both humoral and cellular immunity may be required to prevent congenital infection. Dense bodies (DBs) are complex, noninfectious particles produced by HCMV-infected cells and may represent a vaccine option. As knowledge of the antigenicity and immunogenicity of DB is incomplete, we explored characterization methods and defined DB production methods, followed by systematic evaluation of neutralization and cell-mediated immune responses to the DB material in BALB/c mice. DBs purified from Towne-infected cultures treated with the viral terminase inhibitor 2-bromo-5,6-dichloro-1-beta-d-ribofuranosyl benzimidazole riboside (BDCRB) were characterized by nanoparticle tracking analysis (NTA), two-dimensional fluorescence difference gel electrophoresis (2D-DIGE), immunoblotting, quantitative enzyme-linked immunosorbent assay, and other methods. The humoral and cellular immune responses to DBs were compared to the immunogenicity of glycoprotein B (gB) administered with the adjuvant AddaVax (gB/AddaVax). DBs induced neutralizing antibodies that prevented viral infection of cultured fibroblasts and epithelial cells and robust cell-mediated immune responses to multiple viral proteins, including pp65, gB, and UL48. In contrast, gB/AddaVax failed to induce neutralizing antibodies that prevented infection of epithelial cells, highlighting a critical difference in the humoral responses induced by these vaccine candidates. Our data advance the potential for the DB vaccine approach, demonstrate important immunogenicity properties, and strongly support the further evaluation of DBs as a CMV vaccine candidate.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/imunologia , Células Epiteliais/virologia , Fibroblastos/virologia , Imunidade Celular , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vacinas contra Citomegalovirus/administração & dosagem , Vacinas contra Citomegalovirus/isolamento & purificação , Células Epiteliais/imunologia , Feminino , Fibroblastos/imunologia , Camundongos , Camundongos Endogâmicos BALB C
5.
Virology ; 436(2): 274-83, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23295021

RESUMO

Cytomegaloviruses utilize overlapping and embedded reading frames as a way to efficiently package and express all genes necessary to carry out a complex lifecycle. Murine cytomegalovirus encodes a mitochondrial-localized inhibitor of Bak oligomerization (vIBO) from m41.1, a reading frame that is embedded within the m41 gene. The m41.1-encoded mitochondrial protein and m41-encoded Golgi-localized protein have both been implicated in cell death suppression; however, their contribution to viral infection within the host has not been investigated. Here, we report that mitochondrial-localized m41.1 (vIBO) is required for optimal viral replication in macrophages and has a modest impact on dissemination in infected mice. In contrast, Golgi-localized m41 protein is dispensable during acute infection and dissemination as well as for latency. All together, these data indicate that the primary evolutionary focus of this locus is to maintain mitochondrial function through inhibition of Bak-mediated death pathways in support of viral pathogenesis.


Assuntos
Regulação Viral da Expressão Gênica , Infecções por Herpesviridae/patologia , Muromegalovirus/genética , Muromegalovirus/fisiologia , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Estruturas Animais/virologia , Animais , Sequência de Bases , Modelos Animais de Doenças , Células Epiteliais/virologia , Fibroblastos/virologia , Deleção de Genes , Infecções por Herpesviridae/virologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Muromegalovirus/patogenicidade , Carga Viral , Proteínas Virais/genética , Fatores de Virulência/genética
6.
Virology ; 435(2): 250-7, 2013 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-23159167

RESUMO

At a low MOI (≤0.01), cytomegalovirus-associated programmed cell death terminates productive infection via a pathway triggered by the mitochondrial serine protease HtrA2/Omi. This infected cell death is associated with late phase replication events naturally suppressed by the viral mitochondrial inhibitor of apoptosis (vMIA). Here, higher MOI (ranging from 0.1-3.0) triggers cell death earlier during infection independent of viral DNA synthesis. Thus, MOI-dependent activating signals early, at high MOI, or late, at low MOI, during replication promote serine protease-dependent death that is suppressed by vMIA. Treatment with an antioxidant targeting reactive oxygen species (ROS) or the serine protease inhibitor N-alpha-p-tosyl-L-lysine chloromethyl ketone (TLCK) delays cell death, and the combination has an additive impact. These studies identify serine proteases and ROS as important factors triggering programmed cell death induced by vMIA-deficient virus, and show that this death pathway occurs earlier and reduces viral yields as the MOI is increased.


Assuntos
Morte Celular/efeitos dos fármacos , Citomegalovirus/patogenicidade , Fibroblastos/virologia , Proteínas Mitocondriais/metabolismo , Serina Endopeptidases/metabolismo , Apoptose , Células Cultivadas , Citomegalovirus/fisiologia , Efeito Citopatogênico Viral , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Proteínas Mitocondriais/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Serina Endopeptidases/farmacologia , Inibidores de Serina Proteinase/farmacologia , Proteínas Virais/metabolismo , Replicação Viral
7.
J Virol ; 86(23): 12879-90, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22993151

RESUMO

One common sign of human cytomegalovirus infection is altered liver function. Murine cytomegalovirus strain v70 induces a rapid and severe hepatitis in immunocompetent mice that requires the presence of T cells in order to develop. v70 exhibits approximately 10-fold-greater virulence than the commonly used strain K181, resulting in a more severe, sustained, and lethal hepatitis but not dramatically higher viral replication levels. Hepatitis and death are markedly delayed in immunodeficient SCID compared to immunocompetent BALB/c mice. Transfer of BALB/c splenocytes to SCID mice conferred rapid disease following infection, and depletion of either CD4 or CD8 T cells in BALB/c mice reduced virus-induced hepatitis. The frequency of CD8 T cells producing gamma interferon and tumor necrosis factor in response to viral antigen was higher in settings where more severe disease occurred. Thus, virus-specific effector CD8 T cells appear to contribute to lethal virus-induced hepatitis, contrasting their protective role during sublethal infection. This study reveals how protection and disease during cytomegalovirus infection depend on viral strain and dose, as well as the quality of the T cell response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/complicações , Hepatite Viral Animal/imunologia , Hepatite Viral Animal/virologia , Imunidade Celular/imunologia , Muromegalovirus/patogenicidade , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Citometria de Fluxo , Fluorescência , Hepatite Viral Animal/etiologia , Técnicas Histológicas , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Estatísticas não Paramétricas , Fator de Necrose Tumoral alfa/metabolismo
8.
J Virol ; 84(10): 5108-23, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20219915

RESUMO

The cellular protease caspase-8 activates extrinsic apoptosis and also functions to promote monocyte-to-macrophage differentiation. Differentiation-induced alterations to antiviral caspase-8-dependent cell death pathways are unclear. Here, we show THP-1 monocyte-to-macrophage differentiation alters the specific cell death pathways activated in response to human cytomegalovirus (HCMV) infection. Employing viruses with mutations in UL36, the gene that encodes the viral inhibitor of caspase-8 activation (vICA), our data indicate that both caspase-dependent and -independent death pathways are activated in response to infection. Activation of caspase-dependent and -independent cell death responses restricted growth of vICA-deficient viruses, and vICA/pUL36 inhibited either response. Thus, these studies also reveal that the UL36 gene controls a caspase-independent cell death pathway. The impact of caspases on control of antiviral responses differed at early and late stages of macrophage differentiation. Early in differentiation, vICA-deficient virus-induced cell death was dependent on caspases and inhibited by the pan-caspase inhibitor z-VAD(OMe)-fluoromethyl ketone. In contrast, virus-induced death at late times of differentiation was caspase independent. Additional unlabeled and fluorescent inhibitors indicated that caspase-8 promoted death from within infected cells at early but not late stages of differentiation. These data highlight the multifunctional role of vICA/pUL36 as HCMV encounters various antiviral responses during macrophage differentiation.


Assuntos
Apoptose , Citomegalovirus/patogenicidade , Macrófagos/virologia , Monócitos/virologia , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia , Caspases/metabolismo , Linhagem Celular , Técnicas de Inativação de Genes , Humanos , Proteínas Virais/genética
9.
PLoS Pathog ; 4(5): e1000063, 2008 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-18769594

RESUMO

Viruses encode suppressors of cell death to block intrinsic and extrinsic host-initiated death pathways that reduce viral yield as well as control the termination of infection. Cytomegalovirus (CMV) infection terminates by a caspase-independent cell fragmentation process after an extended period of continuous virus production. The viral mitochondria-localized inhibitor of apoptosis (vMIA; a product of the UL37x1 gene) controls this fragmentation process. UL37x1 mutant virus-infected cells fragment three to four days earlier than cells infected with wt virus. Here, we demonstrate that infected cell death is dependent on serine proteases. We identify mitochondrial serine protease HtrA2/Omi as the initiator of this caspase-independent death pathway. Infected fibroblasts develop susceptibility to death as levels of mitochondria-resident HtrA2/Omi protease increase. Cell death is suppressed by the serine protease inhibitor TLCK as well as by the HtrA2-specific inhibitor UCF-101. Experimental overexpression of HtrA2/Omi, but not a catalytic site mutant of the enzyme, sensitizes infected cells to death that can be blocked by vMIA or protease inhibitors. Uninfected cells are completely resistant to HtrA2/Omi induced death. Thus, in addition to suppression of apoptosis and autophagy, vMIA naturally controls a novel serine protease-dependent CMV-infected cell-specific programmed cell death (cmvPCD) pathway that terminates the CMV replication cycle.


Assuntos
Apoptose , Infecções por Citomegalovirus/patologia , Citomegalovirus/patogenicidade , Proteínas Imediatamente Precoces/fisiologia , Proteínas Mitocondriais/fisiologia , Serina Endopeptidases/fisiologia , Morte Celular , Células Cultivadas , Fibroblastos/patologia , Fibroblastos/virologia , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Humanos , Proteínas Virais/fisiologia , Replicação Viral
10.
J Clin Microbiol ; 45(6): 1804-10, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17409205

RESUMO

Despite antiviral prophylaxis, a high percentage (over 90%) of heart transplant patients experience active cytomegalovirus (CMV) infection, diagnosed by detection of viral DNA in peripheral blood polymorphonuclear leukocytes within the first few months posttransplantation. Viral DNA was detected in mononuclear cells prior to detection in granulocytes from CMV-seropositive recipients (R+) receiving a heart from a CMV-seropositive donor (D+). Based on assessment of systemic infection in leukocyte populations, both R+ subgroups (R+/D- and R+/D+) experienced a greater infection burden than the R-/D+ subgroup, which was aggressively treated because of a higher risk of acute CMV disease. Despite widespread systemic infection in all at-risk patient subgroups, CMV DNA was rarely (< 3% of patients) detected in transplanted heart biopsy specimens. The R+ patients more frequently exceeded the 75th percentile of the CMV DNA copy number distribution in leukocytes (110 copies/10(5) polymorphonuclear leukocytes) than the R-/D+ subgroup. Therefore, active systemic CMV infection involving leukocytes is common in heart transplant recipients receiving prophylaxis to reduce acute disease. Infection of the transplanted organ is rare, suggesting that chronic vascular disease attributed to CMV may be driven by the consequences of systemic infection.


Assuntos
Antivirais/uso terapêutico , Citomegalovirus/isolamento & purificação , Ganciclovir/uso terapêutico , Transplante de Coração/efeitos adversos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Viremia , Adulto , Antivirais/administração & dosagem , Quimioprevenção , Citomegalovirus/genética , Infecções por Citomegalovirus/epidemiologia , Infecções por Citomegalovirus/prevenção & controle , Infecções por Citomegalovirus/virologia , DNA Viral/sangue , Feminino , Ganciclovir/administração & dosagem , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Fatores Imunológicos/administração & dosagem , Incidência , Masculino , Pessoa de Meia-Idade , Viremia/epidemiologia , Viremia/prevenção & controle , Viremia/virologia
11.
J Virol ; 79(19): 12205-17, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16160147

RESUMO

Human cytomegalovirus carries a mitochondria-localized inhibitor of apoptosis (vMIA) that is conserved in primate cytomegaloviruses. We find that inactivating mutations within UL37x1, which encodes vMIA, do not substantially affect replication in TownevarATCC (Towne-BAC), a virus that carries a functional copy of the betaherpesvirus-conserved viral inhibitor of caspase 8 activation, the UL36 gene product. In Towne-BAC infection, vMIA reduces susceptibility of infected cells to intrinsic death induced by proteasome inhibition. vMIA is sufficient to confer resistance to proteasome inhibition when expressed independent of viral infection. Murine cytomegalovirus m38.5, whose position in the viral genome is analogous to UL37x1, exhibits mitochondrial association and functions in much the same manner as vMIA in inhibiting intrinsic cell death. This work suggests a common role for vMIA in rodent and primate cytomegaloviruses, modulating the threshold of virus-infected cells to intrinsic cell death.


Assuntos
Apoptose , Citomegalovirus/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Mitocôndrias/metabolismo , Muromegalovirus/fisiologia , Proteínas Virais/fisiologia , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Citomegalovirus/genética , Humanos , Proteínas Imediatamente Precoces/genética , Membranas Intracelulares/química , Proteínas Mitocondriais/metabolismo , Muromegalovirus/genética , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Proteínas Virais/genética , Replicação Viral
12.
Virology ; 316(2): 221-33, 2003 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-14644605

RESUMO

Human cytomegalovirus (CMV) genes UL36 and UL37 encode viral inhibitor of caspase-8-induced apoptosis (vICA) and viral mitochondria inhibitor of apoptosis (vMIA), respectively. Rhesus macaque CMV homologues, denoted Rh-vICA and Rh-vMIA, were identified and found to suppress apoptosis. One of these functions was conserved in MCMV, encoded by the M36 gene and denoted M-vICA. Conserved regions were compared to domains important to vICA- and vMIA-mediated cell death suppression. The conserved sequences of primate CMV vMIA homologues overlapped with the two known functional domains, providing further evidence supporting a crucial role of vMIA in cell death suppression. RNA blot analyses revealed that expression of murine and rhesus macaque CMV UL36 and UL37 homologues started early and continued through late times of infection. Murine CMV homologues were expressed with alpha (immediate early) kinetics, like human CMV UL36 and UL37, whereas rhesus macaque CMV homologues exhibited beta (delayed early) kinetics. Despite differences in organization and transcriptional regulation, this region appears to carry out a conserved role in cell death suppression. When viewed in light of sequence conservation, a functional vMIA homologue appears to be encoded by every primate CMV, whereas a functional vICA homologue appears to be encoded by all cytomegaloviruses for which sequence data are available.


Assuntos
Apoptose , Chlorocebus aethiops/virologia , Proteínas Imediatamente Precoces/fisiologia , Macaca mulatta/virologia , Muromegalovirus/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Sequência Conservada , Perfilação da Expressão Gênica , Proteínas Imediatamente Precoces/química , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Proteínas Virais/química
13.
J Virol ; 77(1): 631-41, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12477866

RESUMO

By 24 h after infection with human cytomegalovirus, the reticular mitochondrial network characteristic of uninfected fibroblasts was disrupted as mitochondria became punctate and dispersed. These alterations were associated with expression of the immediate-early (alpha) antiapoptotic UL37x1 gene product viral mitochondrion-localized inhibitor of apoptosis (vMIA). Similar alterations in mitochondrial morphology were induced directly by vMIA in transfected cells. A 68-amino-acid antiapoptotic derivative of vMIA containing the mitochondrial localization and antiapoptotic domains also induced disruption, whereas a mutant lacking the antiapoptotic domain failed to cause disruption. These data suggest that the fission and/or fusion process that normally controls mitochondrial networks is altered by vMIA. Mitochondrial fission has been implicated in the induction of apoptosis and vMIA-mediated inhibition of apoptosis may occur subsequent to this event.


Assuntos
Apoptose , Citomegalovirus/patogenicidade , Proteínas Imediatamente Precoces/fisiologia , Mitocôndrias/patologia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Virais , Animais , Células Cultivadas , Humanos , Proteínas Imediatamente Precoces/análise , Proteínas Imediatamente Precoces/química , Camundongos , Mitocôndrias/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...