Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
J Virol ; 93(15)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31092580

RESUMO

Cytomegaloviruses (CMVs) colonize blood-borne myeloid cells. Murine CMV (MCMV) spreads from the lungs via infected CD11c+ cells, consistent with an important role for dendritic cells (DC). We show here that MCMV entering via the olfactory epithelium, a natural transmission portal, also spreads via infected DC. They reached lymph nodes, entered the blood via high endothelial venules, and then entered the salivary glands, driven by constitutive signaling of the viral M33 G protein-coupled receptor (GPCR). Intraperitoneal infection also delivered MCMV to the salivary glands via DC. However, it also seeded F4/80+ infected macrophages to the blood; they did not enter the salivary glands or require M33 for extravasation. Instead, they seeded infection to a range of other sites, including brown adipose tissue (BAT). Peritoneal cells infected ex vivo then adoptively transferred showed similar cell type-dependent differences in distribution, with abundant F4/80+ cells in BAT and CD11c+ cells in the salivary glands. BAT colonization by CMV-infected cells was insensitive to pertussis toxin inhibition of the GPCR signaling through Gi/o substrate, whereas salivary gland colonization was sensitive. Since salivary gland infection required both M33 and Gi/o-coupled signaling, whereas BAT infection required neither, these migrations were mechanistically distinct. MCMV spread from the lungs or nose depended on DC, controlled by M33. Infecting other monocyte populations resulted in unpredictable new infections.IMPORTANCE Cytomegaloviruses (CMVs) spread through the blood by infecting monocytes, and this can lead to disease. With murine CMV (MCMV) we can track infected myeloid cells and so understand how CMVs spread. Previous experiments have injected MCMV into the peritoneal cavity. MCMV normally enters mice via the olfactory epithelium. We show that olfactory infection spreads via dendritic cells, which MCMV directs to the salivary glands. Peritoneal infection similarly reached the salivary glands via dendritic cells. However, it also infected other monocyte types, and they spread infection to other tissues. Thus, infecting the "wrong" monocytes altered virus spread, with potential to cause disease. These results provide a basis for understanding how the monocyte types infected by human CMV might promote different infection outcomes.


Assuntos
Infecções por Citomegalovirus/virologia , Células Dendríticas/virologia , Muromegalovirus/crescimento & desenvolvimento , Células Mieloides/virologia , Estruturas Animais/virologia , Animais , Líquidos Corporais/virologia , Modelos Animais de Doenças , Transmissão de Doença Infecciosa , Humanos , Camundongos
2.
J Neurovirol ; 25(4): 496-507, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31025265

RESUMO

Microglia are resident brain macrophages with key roles in development and brain homeostasis. Cytomegalovirus (CMV) readily infects microglia cells, even as a possible primary target of infection in development. Effects of CMV infection on a cellular level in microglia are still unclear; therefore, the aim of this research was to assess the immunometabolic changes of BV-2 microglia cells following the murine cytomegalovirus (MCMV) infection. In light of that aim, we established an in vitro model of ramified BV-2 microglia (BV-2∅FCS, inducible nitric oxide synthase (iNOSlow), arginase-1 (Arg-1high), mannose receptor CD206high, and hypoxia-inducible factor 1α (HIF-1αlow)) to better replicate the in vivo conditions by removing FCS from the cultivation media, while the cells cultivated in 10% FCS DMEM displayed an ameboid morphology (BV-2FCS high, iNOShigh, Arg-1low, CD206low, and HIF-1αhigh). Experiments were performed using both ramified and ameboid microglia, and both of them were permissive to productive viral infection. Our results indicate that MCMV significantly alters the immunometabolic phenotypic properties of BV-2 microglia cells through the manipulation of iNOS and Arg-1 expression patterns, along with an induction of a glycolytic shift in the infected cell cultures.


Assuntos
Arginase/imunologia , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Microglia/virologia , Muromegalovirus/genética , Óxido Nítrico Sintase Tipo II/imunologia , Animais , Arginase/genética , Linhagem Celular , Meios de Cultura Livres de Soro/farmacologia , Embrião de Mamíferos , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Lectinas Tipo C/deficiência , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Receptor de Manose , Lectinas de Ligação a Manose/deficiência , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Microglia/imunologia , Modelos Biológicos , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/metabolismo , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Cultura Primária de Células , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Transdução de Sinais
3.
Med Microbiol Immunol ; 208(3-4): 543-554, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31115653

RESUMO

Natural killer (NK) cells provide important host defense against herpesvirus infections and influence subsequent T cell control of replication and maintenance of latency. NK cells exhibit phases of expansion, contraction and memory formation in response to the natural mouse pathogen murine cytomegalovirus (MCMV). Innate and adaptive immune responses are tightly regulated in mammals to avoid excess tissue damage while preventing acute and chronic viral disease and assuring resistance to reinfection. Caspase (CASP)8 is an autoactivating aspartate-specific cysteine protease that initiates extrinsic apoptosis and prevents receptor interacting protein (RIP) kinase (RIPK)1-RIPK3-driven necroptosis. CASP8 also promotes death-independent signal transduction. All of these activities make contributions to inflammation. Here, we demonstrate that CASP8 restricts NK cell expansion during MCMV infection but does not influence NK memory. Casp8-/-Ripk3-/- mice mount higher NK response levels than Casp8+/-Ripk3-/- littermate controls or WT C57BL/6 J mice, indicating that RIPK3 deficiency alone does not contribute to NK response patterns. MCMV m157-responsive Ly49H+ NK cells support increased expansion of both Ly49H- NK cells and CD8 T cells in Casp8-/-Ripk3-/- mice. Surprisingly, hyperaccumulation of NK cells depends on the pronecrotic kinase RIPK1. Ripk1-/-Casp8-/-Ripk3-/- mice fail to show the enhanced expansion of lymphocytes observed in Casp8-/-Ripk3-/- mice even though development and homeostasis are preserved in uninfected Ripk1-/-Casp8-/-Ripk3-/- mice. Thus, CASP8 naturally regulates the magnitude of NK cell responses in response to infection where strong activation signals depend on another key regulator of death signaling, RIPK1. In addition, the strong NK cell response promotes survival of effector CD8 T cells during their expansion. Thus, hyperaccumulation of NK cells and crosstalk with T cells becomes amplified in the absence of extrinsic cell death machinery.


Assuntos
Caspase 8/metabolismo , Infecções por Citomegalovirus/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/imunologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL
4.
Med Microbiol Immunol ; 208(3-4): 457-468, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30848361

RESUMO

Natural transmission of cytomegalovirus (CMV) has been difficult to observe. However, recent work using the mouse model of murine (M)CMV demonstrated that MCMV initially infects the nasal mucosa after transmission from mothers to pups. We found that intranasal (i.n.) inoculation of C57BL/6J mice resulted in reliable recovery of replicating virus from the nasal mucosa as assessed by plaque assay. After i.n. inoculation, CD8+ T-cell priming occurred in the mandibular, deep-cervical, and mediastinal lymph nodes within 3 days of infection. Although i.n. infection induced "memory inflation" of T cells specific for the M38316-323 epitope, there were no detectable CD8+ T-cell responses against the late-appearing IE3416-423 epitope, which contrasts with intraperitoneal (i.p.) infection. MCMV-specific T cells migrated into the nasal mucosa where they developed a tissue-resident memory (TRM) phenotype and this could occur independently of local virus infection or antigen. Strikingly however, virus replication was poorly controlled in the nasal mucosa and MCMV was detectable by plaque assay for at least 4 months after primary infection, making the nasal mucosa a second site for MCMV persistence. Unlike in the salivary glands, the persistence of MCMV in the nasal mucosa was not modulated by IL-10. Taken together, our data characterize the development of local and systemic T-cell responses after intranasal infection by MCMV and define the nasal mucosa, a natural site of viral entry, as a novel site of viral persistence.


Assuntos
Infecções por Citomegalovirus/imunologia , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/imunologia , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Linfócitos T/imunologia , Replicação Viral , Animais , Modelos Animais de Doenças , Imunidade Celular , Camundongos Endogâmicos C57BL
5.
Med Microbiol Immunol ; 208(3-4): 555-571, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31098689

RESUMO

Caspase-8 (CASP8) impacts antiviral immunity in expected as well as unexpected ways. Mice with combined deficiency in CASP8 and RIPK3 cannot support extrinsic apoptosis or RIPK3-dependent programmed necrosis, enabling studies of CASP8 function without complications of unleashed necroptosis. These extrinsic cell death pathways are naturally targeted by murine cytomegalovirus (MCMV)-encoded cell death suppressors, showing they are key to cell-autonomous host defense. Remarkably, Casp8-/-Ripk3-/-, Ripk1-/-Casp8-/-Ripk3-/- and Casp8-/-Ripk3K51A/K51A mice mount robust antiviral T cell responses to control MCMV infection. Studies in Casp8-/-Ripk3-/- mice show that CASP8 restrains expansion of MCMV-specific natural killer (NK) and CD8 T cells without compromising contraction or immune memory. Infected Casp8-/-Ripk3-/- or Casp8-/-Ripk3K51A/K51A mice have higher levels of virus-specific NK cells and CD8 T cells compared to matched RIPK3-deficient littermates or WT mice. CASP8, likely acting downstream of Fas death receptor, dampens proliferation of CD8 T cells during expansion. Importantly, contraction proceeds unimpaired in the absence of extrinsic death pathways owing to intact Bim-dependent (intrinsic) apoptosis. CD8 T cell memory develops in Casp8-/-Ripk3-/- mice, but memory inflation characteristic of MCMV infection is not sustained in the absence of CASP8 function. Despite this, Casp8-/-Ripk3-/- mice are immune to secondary challenge. Interferon (IFN)γ is recognized as a key cytokine for adaptive immune control of MCMV. Ifngr-/-Casp8-/-Ripk3-/- mice exhibit increased lifelong persistence in salivary glands as well as lungs compared to Ifngr-/- and Casp8-/-Ripk3-/- mice. Thus, mice deficient in CASP8 and RIPK3 are more dependent on IFNγ mechanisms for sustained T cell immune control of MCMV. Overall, appropriate NK- and T cell immunity to MCMV is dependent on host CASP8 function independent of RIPK3-regulated pathways.


Assuntos
Caspase 8/metabolismo , Infecções por Citomegalovirus/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/imunologia , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout
6.
Med Microbiol Immunol ; 208(3-4): 487-494, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30923899

RESUMO

Cytomegalovirus (CMV) infection is a significant public health problem. Congenital CMV infection is a leading infectious cause of long-term neurodevelopmental sequelae, including mental retardation and sensorineural hearing loss. Immune protection against mouse cytomegalovirus (MCMV) is primarily mediated by NK cells and CD8+ T cells, while CD4+ T cells are not needed for control of MCMV in majority of organs in immunocompetent adult mice. Here, we set out to determine the role of CD4+ T cells upon MCMV infection of newborn mice. We provide evidence that CD4+ T cells are essential for clearance of MCMV infection in brain of neonatal mice and for prevention of recurrence of latent MCMV. In addition, we provide evidence that CD4+ T cells are required for induction and maintenance of tissue-resident memory CD8+ T cells in the brain of mice perinatally infected with MCMV.


Assuntos
Encéfalo/imunologia , Encéfalo/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/imunologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Camundongos
7.
Med Microbiol Immunol ; 208(3-4): 531-542, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31004199

RESUMO

As an immune evasion mechanism, cytomegaloviruses (CMVs) have evolved proteins that interfere with cell surface trafficking of MHC class-I (MHC-I) molecules to tone down recognition by antiviral CD8 T cells. This interference can affect the trafficking of recently peptide-loaded MHC-I from the endoplasmic reticulum to the cell surface, thus modulating the presentation of viral peptides, as well as the recycling of pre-existing cell surface MHC-I, resulting in reduction of the level of overall MHC-I cell surface expression. Murine cytomegalovirus (mCMV) was paradigmatic in that it led to the discovery of this immune evasion strategy of CMVs. Members of its m02-m16 gene family code for type-I transmembrane glycoproteins, proven or predicted, most of which carry cargo sorting motifs in their cytoplasmic, C-terminal tail. For the m06 gene product m06 (gp48), the cargo has been identified as being MHC-I, which is linked by m06 to cellular adapter proteins AP-1A and AP-3A through the dileucine motif EPLARLL. Both APs are involved in trans-Golgi network (TGN) cargo sorting and, based on transfection studies, their engagement by the dileucine motif was proposed to be absolutely required to prevent MHC-I exposure at the cell surface. Here, we have tested this prediction in an infection system with the herein newly described recombinant virus mCMV-m06AA, in which the dileucine motif is destroyed by replacing EPLARLL with EPLARAA. This mutation has a phenotype in that the transition of m06-MHC-I complexes from early endosomes (EE) to late endosomes (LE)/lysosomes for degradation is blocked. Consistent with the binding of the MHC-I α-chain to the luminal domain of m06, the m06-mediated disposal of MHC-I did not require the ß2m chain of mature MHC-I. Unexpectedly, however, disconnecting MHC-I cargo from AP-1A/3A by the motif mutation in m06 had no notable rescuing impact on overall cell surface MHC-I, though it resulted in some improvement of the presentation of viral antigenic peptides by recently peptide-loaded MHC-I. Thus, the current view on the mechanism by which m06 mediates immune evasion needs to be revised. While the cargo sorting motif is critically involved in the disposal of m06-bound MHC-I in the endosomal/lysosomal pathway at the stage of EE to LE transition, this motif-mediated disposal is not the critical step by which m06 causes immune evasion. We rather propose that engagement of AP-1A/3A by the cargo sorting motif in m06 routes the m06-MHC-I complexes into the endosomal pathway and thereby detracts them from the constitutive cell surface transport.


Assuntos
Infecções por Citomegalovirus/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Evasão da Resposta Imune , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/imunologia , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Células Cultivadas , Endossomos/metabolismo , Lisossomos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ligação Proteica , Transporte Proteico
8.
Med Microbiol Immunol ; 208(3-4): 439-446, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31004200

RESUMO

Roizman's definition of herpesviral latency, which applies also to cytomegaloviruses (CMVs), demands maintenance of reactivation-competent viral genomes after clearance of productive infection. It is more recent understanding that failure to complete the productive viral cycle for virus assembly and release does not imply viral gene silencing at all genetic loci and all the time. It rather appears that CMV latency is transcriptionally "noisy" in that silenced viral genes get desilenced from time to time in a stochastic manner, leading to "transcripts expressed in latency" (TELs). If a TEL happens to code for a protein that contains a CD8 T cell epitope, protein processing can lead to the presentation of the antigenic peptide and restimulation of cognate CD8 T cells during latency. This mechanism is discussed as a potential driver of epitope-selective accumulation of CD8 T cells over time, a phenomenon linked to CMV latency and known as "memory inflation" (MI). So far, expression of an epitope-encoding TEL was shown only for the major immediate-early (MIE) gene m123/ie1 of murine cytomegalovirus (mCMV), which codes for the prototypic MI-driving antigenic peptide YPHFMPTNL that is presented by the MHC class-I molecule Ld. The only known second MI-driving antigenic peptide of mCMV in the murine MHC haplotype H-2d is AGPPRYSRI presented by the MHC-I molecule Dd. This peptide is very special in that it is encoded by the early (E) phase gene m164 and by an overlapping immediate-early (IE) transcript governed by a promoter upstream of m164. If MI is driven by presentation of TEL-derived antigenic peptides, as the hypothesis says, one should find corresponding TELs. We show here that E-phase and IE-phase transcripts that code for the MI-driving antigenic peptide AGPPRYSRI are independently and stochastically expressed in latently infected lungs.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Perfilação da Expressão Gênica , Muromegalovirus/imunologia , Latência Viral , Animais , Antígenos Virais/biossíntese , Modelos Animais de Doenças , Epitopos/biossíntese , Epitopos/imunologia , Memória Imunológica , Muromegalovirus/crescimento & desenvolvimento
9.
J Gen Virol ; 99(11): 1509-1514, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30226462

RESUMO

Human cytomegalovirus (HCMV) colonizes blood-borne dendritic cells (DCs). They express US28, a viral G protein-coupled receptor (GPCR). In vitro functions have been described for US28, but how it contributes to host colonization has been unclear. The murine CMV (MCMV) M33 GPCR promotes DC recirculation. We show that US28 shares this function. Thus, DC recirculation is also available to HCMV via US28, and inhibiting US28 G protein-dependent signalling has the potential to reduce systemic infection. We show that M33 also promotes systemic infection through infected DC extravasation.


Assuntos
Movimento Celular , Infecções por Citomegalovirus/virologia , Citomegalovirus/patogenicidade , Células Dendríticas/virologia , Interações Hospedeiro-Patógeno , Linfonodos/virologia , Receptores de Quimiocinas/metabolismo , Proteínas Virais/metabolismo , Estruturas Animais/virologia , Animais , Células Cultivadas , Citomegalovirus/crescimento & desenvolvimento , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/veterinária , Células Dendríticas/imunologia , Humanos , Linfonodos/imunologia , Camundongos Endogâmicos BALB C , Muromegalovirus/crescimento & desenvolvimento
10.
Mol Vis ; 24: 379-394, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29853772

RESUMO

Purpose: The purpose of this study was to determine whether the blood-retina barrier is compromised by choroidal murine cytomegalovirus (MCMV) infection, using electron microscopy. Methods: BALB/c mice were immunosuppressed with methylprednisolone and monoclonal antibodies to CD4 and CD8. At several time points post-MCMV intraperitoneal inoculation, the eyes were removed and analyzed with western blotting and immunoelectron microscopy for the presence of MCMV early antigen (EA) and the host protein RIP3. Posterior eyecups from RIP3-/- and RIP3+/+ mice were cultured and inoculated with MCMV. At days 4, 7, and 11 post-infection, cultures were collected and analyzed with plaque assay, immunohistochemical staining, and real-time PCR (RT-PCR). Results: MCMV EA was observed in the nuclei of vascular endothelial cells and pericytes in the choriocapillaris. Disruption of Bruch's membrane was observed, especially at sites adjacent to activated platelets, and a few RPE cells containing some enlarged vesicles were found directly beneath disrupted Bruch's membrane. Some virus particles were also observed in the enlarged vesicles of RPE cells. Levels of the RIP3 protein, which was observed mainly in the RPE cells and the basement membrane of the choriocapillaris, were greatly increased following MCMV infection, while depletion of RIP3 resulted in greatly decreased inflammasome formation, as well as expression of downstream inflammation factors. Conclusions: The results suggest that systemic MCMV spreads to the choroid and replicates in vascular endothelia and pericytes of the choriocapillaris during immunosuppression. Choroidal MCMV infection is associated with in situ inflammation and subsequent disruption of Bruch's membrane and the outer blood-retina barrier.


Assuntos
Corioide/imunologia , Infecções por Citomegalovirus/imunologia , Infecções Oculares Virais/imunologia , Hospedeiro Imunocomprometido , Retina/imunologia , Retinite/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Antígenos Virais/genética , Plaquetas/imunologia , Plaquetas/patologia , Plaquetas/virologia , Barreira Hematorretiniana/imunologia , Barreira Hematorretiniana/patologia , Barreira Hematorretiniana/virologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Corioide/irrigação sanguínea , Corioide/patologia , Corioide/virologia , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Células Endoteliais , Infecções Oculares Virais/patologia , Infecções Oculares Virais/virologia , Feminino , Proteínas Imediatamente Precoces/genética , Inflamassomos/imunologia , Metilprednisolona/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/patogenicidade , Pericitos/imunologia , Pericitos/patologia , Pericitos/virologia , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Retina/patologia , Retina/virologia , Epitélio Pigmentado da Retina/imunologia , Epitélio Pigmentado da Retina/patologia , Epitélio Pigmentado da Retina/virologia , Retinite/patologia , Retinite/virologia
12.
J Gen Virol ; 96(Pt 5): 1127-1137, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25575707

RESUMO

Infection with multiple genetically distinct strains of pathogen is common and can lead to positive (complementation) or negative (competitive) within-host interactions. These interactions can alter aspects of the disease process and help shape pathogen evolution. Infection of the host with multiple strains of cytomegalovirus (CMV) occurs frequently in humans and mice. Profound, NK-cell-mediated (apparent) competition has been identified in C57BL/6 mice, and prevented the replication and shedding of certain co-infecting CMV strains. However, the frequency of such strong competition has not been established. Other within-host interactions such as complementation or alternative forms of competition remain possible. Moreover, high rates of recombination in both human CMV and murine CMV (MCMV) suggest prolonged periods of viral co-replication, rather than strong competitive suppression. An established model was employed to investigate the different possible outcomes of multi-strain infection in other mouse strains. In this study, co-replication of up to four strains of MCMV in the spleen, liver and salivary glands was observed in both MCMV-susceptible and MCMV-resistant mice. In the absence of apparent competition, no other forms of competition were unmasked. In addition, no evidence of complementation between viral strains was observed. Importantly, co-replication of MCMV strains was apparent for up to 90 days in the salivary glands. These data indicated that competition was not the default outcome of multi-strain CMV infection. Prolonged, essentially neutral, co-replication may be the norm, allowing for multi-strain transmission and prolonged opportunities for recombination.


Assuntos
Coinfecção/virologia , Infecções por Herpesviridae/virologia , Muromegalovirus/crescimento & desenvolvimento , Subfamília A de Receptores Semelhantes a Lectina de Células NK/imunologia , Glândulas Salivares/virologia , Animais , Fígado/virologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Subfamília A de Receptores Semelhantes a Lectina de Células NK/deficiência , Baço/virologia
13.
PLoS Pathog ; 9(12): e1003828, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24348257

RESUMO

Neonates, including mice and humans, are highly susceptible to cytomegalovirus (CMV) infection. However, many aspects of neonatal CMV infections such as viral cell tropism, spatio-temporal distribution of the pathogen as well as genesis of antiviral immunity are unknown. With the use of reporter mutants of the murine cytomegalovirus (MCMV) we identified the lung as a primary target of mucosal infection in neonatal mice. Comparative analysis of neonatal and adult mice revealed a delayed control of virus replication in the neonatal lung mucosa explaining the pronounced systemic infection and disease in neonates. This phenomenon was supplemented by a delayed expansion of CD8(+) T cell clones recognizing the viral protein M45 in neonates. We detected viral infection at the single-cell level and observed myeloid cells forming "nodular inflammatory foci" (NIF) in the neonatal lung. Co-localization of infected cells within NIFs was associated with their disruption and clearance of the infection. By 2-photon microscopy, we characterized how neonatal antigen-presenting cells (APC) interacted with T cells and induced mature adaptive immune responses within such NIFs. We thus define NIFs of the neonatal lung as niches for prolonged MCMV replication and T cell priming but also as sites of infection control.


Assuntos
Infecções por Citomegalovirus/imunologia , Pulmão/imunologia , Muromegalovirus/imunologia , Pneumonia/imunologia , Pneumonia/virologia , Linfócitos T/imunologia , Animais , Animais Recém-Nascidos , Apresentação de Antígeno , Células Cultivadas , Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Intestinos/imunologia , Intestinos/patologia , Intestinos/virologia , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Pulmão/virologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Muromegalovirus/crescimento & desenvolvimento , Pneumonia/patologia
14.
PLoS Pathog ; 8(8): e1002901, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952450

RESUMO

Little is known about the role of viral genes in modulating host cytokine responses. Here we report a new functional role of the viral encoded IE1 protein of the murine cytomegalovirus in sculpting the inflammatory response in an acute infection. In time course experiments of infected primary macrophages (MΦs) measuring cytokine production levels, genetic ablation of the immediate-early 1 (ie1) gene results in a significant increase in TNFα production. Intracellular staining for cytokine production and viral early gene expression shows that TNFα production is highly associated with the productively infected MΦ population of cells. The ie1- dependent phenotype of enhanced MΦ TNFα production occurs at both protein and RNA levels. Noticeably, we show in a series of in vivo infection experiments that in multiple organs the presence of ie1 potently inhibits the pro-inflammatory cytokine response. From these experiments, levels of TNFα, and to a lesser extent IFNß, but not the anti-inflammatory cytokine IL10, are moderated in the presence of ie1. The ie1- mediated inhibition of TNFα production has a similar quantitative phenotype profile in infection of susceptible (BALB/c) and resistant (C57BL/6) mouse strains as well as in a severe immuno-ablative model of infection. In vitro experiments with infected macrophages reveal that deletion of ie1 results in increased sensitivity of viral replication to TNFα inhibition. However, in vivo infection studies show that genetic ablation of TNFα or TNFRp55 receptor is not sufficient to rescue the restricted replication phenotype of the ie1 mutant virus. These results provide, for the first time, evidence for a role of IE1 as a regulator of the pro-inflammatory response and demonstrate a specific pathogen gene capable of moderating the host production of TNFα in vivo.


Assuntos
Regulação Viral da Expressão Gênica/genética , Infecções por Herpesviridae/imunologia , Proteínas Imediatamente Precoces/genética , Muromegalovirus/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Replicação do DNA , DNA Viral/genética , Feminino , Infecções por Herpesviridae/virologia , Proteínas Imediatamente Precoces/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/fisiologia , Fenótipo , Transdução de Sinais , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
15.
Proc Natl Acad Sci U S A ; 108(38): 15978-83, 2011 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-21930925

RESUMO

Antiviral compounds that increase the resistance of host tissues represent an attractive class of therapeutic. Here, we show that squalamine, a compound previously isolated from the tissues of the dogfish shark (Squalus acanthias) and the sea lamprey (Petromyzon marinus), exhibits broad-spectrum antiviral activity against human pathogens, which were studied in vitro as well as in vivo. Both RNA- and DNA-enveloped viruses are shown to be susceptible. The proposed mechanism involves the capacity of squalamine, a cationic amphipathic sterol, to neutralize the negative electrostatic surface charge of intracellular membranes in a way that renders the cell less effective in supporting viral replication. Because squalamine can be readily synthesized and has a known safety profile in man, we believe its potential as a broad-spectrum human antiviral agent should be explored.


Assuntos
Antivirais/farmacologia , Viroses/tratamento farmacológico , Replicação Viral/efeitos dos fármacos , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Antivirais/química , Linhagem Celular , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Colestanóis/química , Colestanóis/farmacologia , Cricetinae , Feminino , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/crescimento & desenvolvimento , Vírus Delta da Hepatite/efeitos dos fármacos , Vírus Delta da Hepatite/crescimento & desenvolvimento , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Muromegalovirus/efeitos dos fármacos , Muromegalovirus/crescimento & desenvolvimento , Espalhamento a Baixo Ângulo , Viroses/virologia , Difração de Raios X , Proteínas rac1 de Ligação ao GTP/química
16.
J Virol ; 86(23): 12512-24, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22993161

RESUMO

Dominant-negative (DN) mutants are powerful tools for studying essential protein-protein interactions. A systematic genetic screen of the essential murine cytomegalovirus (MCMV) protein pM53 identified the accumulation of inhibitory mutations within conserved region 2 (CR2) and CR4. The strong inhibitory potential of these CR4 mutants is characterized by a particular phenotype. The DN effect of the small insertion mutations in CR2 was too weak to analyze (M. Popa, Z. Ruzsics, M. Lötzerich, L. Dölken, C. Buser, P. Walther, and U. H. Koszinowski, J. Virol. 84:9035-9046, 2010); therefore, the present study describes the construction of M53 alleles lacking CR2 (either completely or partially) and subsequent examination of the DN effect on MCMV replication upon conditional expression. Overexpression of CR2-deficient pM53 inhibited virus production by about 10,000-fold. This was due to interference with capsid export from the nucleus and viral genome cleavage/packaging. In addition, the fate of the nuclear envelopment complex in the presence of DN pM53 overexpression was analyzed. The CR2 mutants were able to bind to pM50, albeit to a lesser extent than the wild-type protein, and relocalized the wild-type nuclear envelope complex in infected cells. Unlike the CR4 DN, the CR2 DN mutants did not affect the stability of pM50.


Assuntos
Proteínas do Capsídeo/genética , Muromegalovirus/genética , Membrana Nuclear/virologia , Proteínas Nucleares/genética , Replicação Viral/genética , Alelos , Animais , Southern Blotting , Western Blotting , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Teste de Complementação Genética , Imunoprecipitação , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Muromegalovirus/crescimento & desenvolvimento , Mutação/genética , Proteínas Nucleares/metabolismo , Plasmídeos/genética , Reação em Cadeia da Polimerase
17.
J Gen Virol ; 93(Pt 7): 1537-1547, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22442111

RESUMO

One goal of virus infection is to reprogramme the host cell to optimize virus replication. As part of this process, viral microRNAs (miRNAs) may compete for components of the miRNA/small interfering RNA pathway, as well as regulate cellular targets. Murine cytomegalovirus (MCMV) has been described to generate large numbers of viral miRNAs during lytic infection and was therefore used to analyse the impact of viral miRNAs on the host-cell small-RNA system, as well as to check for sorting of viral small RNAs into specific Argonaute (Ago) proteins. Deep-sequencing analysis of MCMV-infected cells revealed that viral miRNAs represented only ~13% of all detected miRNAs. All previously described MCMV miRNAs with the exception of miR-m88-1* were confirmed, and for the MCMV miR-m01-1 hairpin, an additional miRNA, designated miR-m01-1-3p, was found. Its presence was confirmed by quantitative real-time PCR and Northern blotting. Deep sequencing after RNA-induced silencing complex (RISC) immunoprecipitation with antibodies specific for either Ago1 or Ago2 showed that all MCMV miRNAs were loaded into both RISCs. The ratio of MCMV to mouse miRNAs was not increased after immunoprecipitation of Ago proteins. Viral miRNAs therefore did not overwhelm the host miRNA processing system, nor were they incorporated preferentially into RISCs. Three mouse miRNAs were found that showed altered expression as a result of MCMV infection. Downregulation of miR-27a, as described previously, could be confirmed. In addition, miR-26a was downregulated, and upregulation of miR-7a dependent on viral protein expression could be observed.


Assuntos
Expressão Gênica , Interações Hospedeiro-Patógeno , MicroRNAs/biossíntese , Muromegalovirus/crescimento & desenvolvimento , Animais , Northern Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real
18.
J Virol ; 85(19): 10286-99, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21775459

RESUMO

Activated macrophages play a central role in controlling inflammatory responses to infection and are tightly regulated to rapidly mount responses to infectious challenge. Type I interferon (alpha/beta interferon [IFN-α/ß]) and type II interferon (IFN-γ) play a crucial role in activating macrophages and subsequently restricting viral infections. Both types of IFNs signal through related but distinct signaling pathways, inducing a vast number of interferon-stimulated genes that are overlapping but distinguishable. The exact mechanism by which IFNs, particularly IFN-γ, inhibit DNA viruses such as cytomegalovirus (CMV) is still not fully understood. Here, we investigate the antiviral state developed in macrophages upon reversible inhibition of murine CMV by IFN-γ. On the basis of molecular profiling of the reversible inhibition, we identify a significant contribution of a restricted type I IFN subnetwork linked with IFN-γ activation. Genetic knockout of the type I-signaling pathway, in the context of IFN-γ stimulation, revealed an essential requirement for a primed type I-signaling process in developing a full refractory state in macrophages. A minimal transient induction of IFN-ß upon macrophage activation with IFN-γ is also detectable. In dose and kinetic viral replication inhibition experiments with IFN-γ, the establishment of an antiviral effect is demonstrated to occur within the first hours of infection. We show that the inhibitory mechanisms at these very early times involve a blockade of the viral major immediate-early promoter activity. Altogether our results show that a primed type I IFN subnetwork contributes to an immediate-early antiviral state induced by type II IFN activation of macrophages, with a potential further amplification loop contributed by transient induction of IFN-ß.


Assuntos
Interferon Tipo I/imunologia , Interferon gama/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/imunologia , Animais , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transdução de Sinais , Fatores de Tempo
19.
J Med Virol ; 84(6): 912-22, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22499015

RESUMO

A mutant of murine cytomegalovirus (MCMV), tsm5, which is temperature-sensitive for replication in murine embryo fibroblasts at 40°C, failed to replicate to detectable levels in mice. A total of 18 non-synonymous mutations have been identified in tsm5. In a previous study, a mutation (C890Y) identified in the M70 primase gene, when introduced into the wt M70 primase, resulted in a mutant with reduced viral replication at 40°C in vitro and which was severely attenuated in vivo. Five other previously identified mutations may also contribute to the tsm5 phenotype: (1) an A658S mutation in a protein expressed by the M27 ORF; (2) a V54I mutation in M36; (3) a Y565* mutation in m139; (4) a V195M mutation in m141; and (5) an M232I mutation in m143. In the present study, the above-mentioned mutations were introduced individually (M27, M36, m139, m141, m143) or together (M27/M36) into the MCMV K181 (Perth) variant bacterial artificial chromosome (BAC) using RecE/T homologous recombination. Growth in culture revealed that, apart from the double mutant (M27 and M36) and the m139 mutant, the introduced mutations in the above-mentioned genes did not show a temperature-sensitive phenotype in MEF or Raw 264.7 macrophage cells compared to their revertants or the wt virus. In contrast, replication of the M27/M36 double mutant was drastically reduced in MEFs at 40°C and in macrophages at 37°C. Replication of the m139 mutant was reduced in MEF cells at 40°C but not in macrophages. Thus, at least three further mutations contribute to the tsm5 phenotype.


Assuntos
Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/genética , Mutação de Sentido Incorreto , Fases de Leitura Aberta , Replicação Viral/efeitos da radiação , Animais , Células Cultivadas , DNA Viral/química , DNA Viral/genética , Fibroblastos/virologia , Masculino , Camundongos , Muromegalovirus/efeitos da radiação , Fenótipo , Análise de Sequência de DNA , Temperatura
20.
J Virol ; 84(16): 7994-8006, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519391

RESUMO

Cytomegaloviruses, representatives of the Betaherpesvirinae, cause opportunistic infections in immunocompromised hosts. They infect various cells and tissues in their natural host but are highly species specific. For instance, human cytomegalovirus (HCMV) does not replicate in mouse cells, and human cells are not permissive for murine cytomegalovirus (MCMV) infection. However, the underlying molecular mechanisms are so far poorly understood. In the present study we isolated and characterized a spontaneously occurring MCMV mutant that has gained the capacity to replicate rapidly and to high titers in human cells. Compared to the parental wild-type (wt) virus, this mutant formed larger nuclear replication compartments and replicated viral DNA more efficiently. It also disrupted promyelocytic leukemia (PML) protein nuclear domains with greater efficiency but caused less apoptosis than did wt MCMV. Sequence analysis of the mutant virus genome revealed mutations in the M112/M113-coding region. This region is homologous to the HCMV UL112-113 region and encodes the viral early 1 (E1) proteins, which are known to play an important role in viral DNA replication. By introducing the M112/M113 mutations into wt MCMV, we demonstrated that they are sufficient to facilitate MCMV replication in human cells and are, at least in part, responsible for the efficient replication capability of the spontaneously adapted virus. However, additional mutations probably contribute as well. These results reveal a previously unrecognized role of the viral E1 proteins in regulating viral replication in different cells and provide new insights into the mechanisms of the species specificity of cytomegaloviruses.


Assuntos
Antígenos Virais/genética , Proteínas Imediatamente Precoces/genética , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/genética , Mutação de Sentido Incorreto , Replicação Viral , Animais , Sobrevivência Celular , Células Cultivadas , Análise Mutacional de DNA , DNA Viral/química , DNA Viral/genética , Humanos , Camundongos , Análise de Sequência de DNA , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA