Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Eur J Immunol ; 52(6): 936-945, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35304741

RESUMO

COVID-19 vaccines prevent severe forms of the disease, but do not warrant complete protection against breakthrough infections. This could be due to suboptimal mucosal immunity at the site of virus entry, given that all currently approved vaccines are administered via the intramuscular route. In this study, we assessed humoral and cellular immune responses in BALB/c mice after intranasal and intramuscular immunization with adenoviral vector ChAdOx1-S expressing full-length Spike protein of SARS-CoV-2. We showed that both routes of vaccination induced a potent IgG antibody response, as well as robust neutralizing capacity, but intranasal vaccination elicited a superior IgA antibody titer in the sera and in the respiratory mucosa. Bronchoalveolar lavage from intranasally immunized mice efficiently neutralized SARS-CoV-2, which has not been the case in intramuscularly immunized group. Moreover, substantially higher percentages of epitope-specific CD8 T cells exhibiting a tissue resident phenotype were found in the lungs of intranasally immunized animals. Finally, both intranasal and intramuscular vaccination with ChAdOx1-S efficiently protected the mice after the challenge with recombinant herpesvirus expressing the Spike protein. Our results demonstrate that intranasal application of adenoviral vector ChAdOx1-S induces superior mucosal immunity and therefore could be a promising strategy for putting the COVID-19 pandemic under control.


Assuntos
COVID-19 , Vacinas Virais , Adenoviridae/genética , Administração Intranasal , Animais , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Imunidade Celular , Imunidade nas Mucosas , Camundongos , Camundongos Endogâmicos BALB C , Pandemias/prevenção & controle , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Vacinação/métodos
2.
Life (Basel) ; 11(10)2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34685468

RESUMO

The twentieth century witnessed a huge expansion in the number of vaccines used with great success in combating diseases, especially the ones caused by viral and bacterial pathogens. Despite this, several major public health threats, such as HIV, tuberculosis, malaria, and cancer, still pose an enormous humanitarian and economic burden. As vaccines based on the induction of protective, neutralizing antibodies have not managed to effectively combat these diseases, in recent decades, the focus has increasingly shifted towards the cellular immune response. There is substantial evidence demonstrating CD8 T cells as key players in the protection not only against many viral and bacterial pathogens, but also in the fight against neoplastic cells. Here, we present arguments for CD8 T cells to be considered as promising candidates for vaccine targeting. We discuss the heterogeneity of CD8 T cell populations and their contribution in the protection of the host. We also outline several strategies of using a common human pathogen, cytomegalovirus, as a vaccine vector since accumulated data strongly suggest it represents a promising approach to the development of novel vaccines against both pathogens and tumors.

3.
Front Immunol ; 12: 681380, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34168650

RESUMO

Viral vectors have emerged as a promising alternative to classical vaccines due to their great potential for induction of a potent cellular and humoral immunity. Cytomegalovirus (CMV) is an attractive vaccine vector due to its large genome with many non-essential immunoregulatory genes that can be easily manipulated to modify the immune response. CMV generates a strong antigen-specific CD8 T cell response with a gradual accumulation of these cells in the process called memory inflation. In our previous work, we have constructed a mouse CMV vector expressing NKG2D ligand RAE-1γ in place of its viral inhibitor m152 (RAE-1γMCMV), which proved to be highly attenuated in vivo. Despite attenuation, RAE-1γMCMV induced a substantially stronger CD8 T cell response to vectored antigen than the control vector and provided superior protection against bacterial and tumor challenge. In the present study, we confirmed the enhanced protective capacity of RAE-1γMCMV as a tumor vaccine vector and determined the phenotypical and functional characteristics of memory CD8 T cells induced by the RAE-1γ expressing MCMV. RNAseq data revealed higher transcription of numerous genes associated with effector-like CD8 T cell phenotype in RAE-1γMCMV immunized mice. CD8 T cells primed with RAE-1γMCMV were enriched in TCF1 negative population, with higher expression of KLRG1 and lower expression of CD127, CD27, and Eomes. These phenotypical differences were associated with distinct functional features as cells primed with RAE-1γMCMV showed inferior cytokine-producing abilities but comparable cytotoxic potential. After adoptive transfer into naive hosts, OT-1 cells induced with both RAE-1γMCMV and the control vector were equally efficient in rejecting established tumors, suggesting the context of latent infection and cell numbers as important determinants of enhanced anti-tumor response following RAE-1γMCMV vaccination. Overall, our results shed new light on the phenotypical and functional distinctness of memory CD8 T cells induced with CMV vector expressing cellular ligand for the NKG2D receptor.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas contra Citomegalovirus/imunologia , Memória Imunológica , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Fenótipo , Animais , Vacinas Anticâncer/imunologia , Biologia Computacional/métodos , Citomegalovirus/imunologia , Citotoxicidade Imunológica , Perfilação da Expressão Gênica , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Imunofenotipagem , Ativação Linfocitária/imunologia , Camundongos , Muromegalovirus/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Transcriptoma
4.
Immunity ; 54(7): 1478-1493.e6, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34015257

RESUMO

Viral infections during pregnancy are a considerable cause of adverse outcomes and birth defects, and the underlying mechanisms are poorly understood. Among those, cytomegalovirus (CMV) infection stands out as the most common intrauterine infection in humans, putatively causing early pregnancy loss. We employed murine CMV as a model to study the consequences of viral infection on pregnancy outcome and fertility maintenance. Even though pregnant mice successfully controlled CMV infection, we observed highly selective, strong infection of corpus luteum (CL) cells in their ovaries. High infection densities indicated complete failure of immune control in CL cells, resulting in progesterone insufficiency and pregnancy loss. An abundance of gap junctions, absence of vasculature, strong type I interferon (IFN) responses, and interaction of innate immune cells fully protected the ovarian follicles from viral infection. Our work provides fundamental insights into the effect of CMV infection on pregnancy loss and mechanisms protecting fertility.


Assuntos
Corpo Lúteo/imunologia , Infecções por Citomegalovirus/imunologia , Fertilidade/imunologia , Imunidade Inata/imunologia , Animais , Corpo Lúteo/virologia , Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Feminino , Junções Comunicantes/imunologia , Interferon Tipo I/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Gravidez , Progesterona/imunologia
5.
PLoS Pathog ; 15(9): e1008036, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31525249

RESUMO

Cytomegalovirus (CMV) is a ubiquitous ß-herpesvirus that establishes life-long latent infection in a high percentage of the population worldwide. CMV induces the strongest and most durable CD8+ T cell response known in human clinical medicine. Due to its unique properties, the virus represents a promising candidate vaccine vector for the induction of persistent cellular immunity. To take advantage of this, we constructed a recombinant murine CMV (MCMV) expressing an MHC-I restricted epitope from influenza A virus (IAV) H1N1 within the immediate early 2 (ie2) gene. Only mice that were immunized intranasally (i.n.) were capable of controlling IAV infection, despite the greater potency of the intraperitoneally (i.p.) vaccination in inducing a systemic IAV-specific CD8+ T cell response. The protective capacity of the i.n. immunization was associated with its ability to induce IAV-specific tissue-resident memory CD8+ T (CD8TRM) cells in the lungs. Our data demonstrate that the protective effect exerted by the i.n. immunization was critically mediated by antigen-specific CD8+ T cells. CD8TRM cells promoted the induction of IFNγ and chemokines that facilitate the recruitment of antigen-specific CD8+ T cells to the lungs. Overall, our results showed that locally applied MCMV vectors could induce mucosal immunity at sites of entry, providing superior immune protection against respiratory infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade nas Mucosas , Vacinas contra Influenza/imunologia , Muromegalovirus/imunologia , Administração Intranasal , Sequência de Aminoácidos , Animais , Linhagem Celular , Quimiocinas/biossíntese , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Feminino , Produtos do Gene env/administração & dosagem , Produtos do Gene env/genética , Produtos do Gene env/imunologia , Vetores Genéticos , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/genética , Células NIH 3T3 , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
6.
J Exp Med ; 216(8): 1809-1827, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31142589

RESUMO

CMVs efficiently target MHC I molecules to avoid recognition by cytotoxic T cells. However, the lack of MHC I on the cell surface renders the infected cell susceptible to NK cell killing upon missing self recognition. To counter this, mouse CMV (MCMV) rescues some MHC I molecules to engage inhibitory Ly49 receptors. Here we identify a new viral protein, MATp1, that is essential for MHC I surface rescue. Rescued altered-self MHC I molecules show increased affinity to inhibitory Ly49 receptors, resulting in inhibition of NK cells despite substantially reduced MHC I surface levels. This enables the virus to evade recognition by licensed NK cells. During evolution, this novel viral immune evasion mechanism could have prompted the development of activating NK cell receptors that are specific for MATp1-modified altered-self MHC I molecules. Our study solves a long-standing conundrum of how MCMV avoids recognition by NK cells, unravels a fundamental new viral immune evasion mechanism, and demonstrates how this forced the evolution of virus-specific activating MHC I-restricted Ly49 receptors.


Assuntos
Infecções por Herpesviridae/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Evasão da Resposta Imune/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/metabolismo , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas Virais/metabolismo , Animais , Antígenos Ly/genética , Citotoxicidade Imunológica , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Infecções por Herpesviridae/virologia , Antígenos de Histocompatibilidade Classe I/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor 1 Desencadeador da Citotoxicidade Natural/genética
7.
Front Immunol ; 9: 991, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867968

RESUMO

The development of a vaccine against human cytomegalovirus (CMV) has been a subject of long-term medical interest. The research during recent years identified CMV as an attractive vaccine vector against infectious diseases and tumors. The immune response to CMV persists over a lifetime and its unique feature is the inflationary T cell response to certain viral epitopes. CMV encodes numerous genes involved in immunoevasion, which are non-essential for virus growth in vitro. The deletion of those genes results in virus attenuation in vivo, which enables us to dramatically manipulate its virulence and the immune response. We have previously shown that the murine CMV (MCMV) expressing RAE-1γ, one of the cellular ligands for the NKG2D receptor, is highly attenuated in vivo but retains the ability to induce a strong CD8+ T cell response. Here, we demonstrate that recombinant MCMV expressing high affinity NKG2D ligand murine UL16 binding protein-like transcript (MULT-1) (MULT-1MCMV) inserted in the place of its viral inhibitor is dramatically attenuated in vivo in a NK cell-dependent manner, both in immunocompetent adult mice and in immunologically immature newborns. MULT-1MCMV was more attenuated than the recombinant virus expressing RAE-1γ. Despite the drastic sensitivity to innate immune control, MULT-1MCMV induced an efficient CD8+ T cell response to viral and vectored antigens. By using in vitro assay, we showed that similar to RAE-1γMCMV, MULT-1 expressing virus provided strong priming of CD8+ T cells. Moreover, MULT-1MCMV was able to induce anti-viral antibodies, which after passing the transplacental barrier protect offspring of immunized mothers from challenge infection. Altogether, this study further supports the concept that CMV expressing NKG2D ligand possesses excellent characteristics to serve as a vaccine or vaccine vector.


Assuntos
Proteínas de Transporte/genética , Infecções por Citomegalovirus/imunologia , Vacinas contra Citomegalovirus/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Muromegalovirus/genética , Animais , Animais Recém-Nascidos , Linfócitos T CD8-Positivos/imunologia , Proteínas de Transporte/imunologia , Vacinas contra Citomegalovirus/genética , Modelos Animais de Doenças , Feminino , Vetores Genéticos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunidade Materno-Adquirida , Imunocompetência , Células Matadoras Naturais/imunologia , Proteínas de Membrana , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Muromegalovirus/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia
8.
Eur J Immunol ; 47(8): 1354-1367, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28612942

RESUMO

Designing CD8+ T-cell vaccines, which would provide protection against tumors is still considered a great challenge in immunotherapy. Here we show the robust potential of cytomegalovirus (CMV) vector expressing the NKG2D ligand RAE-1γ as CD8+ T cell-based vaccine against malignant tumors. Immunization with the CMV vector expressing RAE-1γ, delayed tumor growth or even provided complete protection against tumor challenge in both prophylactic and therapeutic settings. Moreover, a potent tumor control in mice vaccinated with this vector can be further enhanced by blocking the immune checkpoints TIGIT and PD-1. CMV vector expressing RAE-1γ potentiated expansion of KLRG1+ CD8+ T cells with enhanced effector properties. This vaccination was even more efficient in neonatal mice, resulting in the expansion and long-term maintenance of epitope-specific CD8+ T cells conferring robust resistance against tumor challenge. Our data show that immunomodulation of CD8+ T-cell responses promoted by herpesvirus expressing a ligand for NKG2D receptor can provide a powerful platform for the prevention and treatment of CD8+ T-cell sensitive tumors.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Citomegalovirus/genética , Proteínas de Membrana/genética , Neoplasias/imunologia , Animais , Animais Recém-Nascidos , Citomegalovirus/imunologia , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Vetores Genéticos , Humanos , Imunomodulação , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Lectinas Tipo C , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Proteínas de Membrana/imunologia , Camundongos , Neoplasias/prevenção & controle , Neoplasias/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia
9.
Immunol Lett ; 189: 40-47, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28414184

RESUMO

Cytomegaloviruses (CMVs) have dedicated a large portion of their genome towards immune evasion targeting many aspects of the host immune system, particularly NK cells. However, the host managed to cope with the infection by developing multiple mechanisms to recognize viral threat and counterattack it, thus illustrating never-ending evolutionary interplay between CMV and its host. In this review, we will focus on several mechanisms of NK cell evasion by mouse CMV (MCMV), the role of host inhibitory and activating Ly49 receptors involved in the virus control and acquisition of adaptive features by NK cells as a consequence of MCMV infection.


Assuntos
Infecções por Herpesviridae/imunologia , Muromegalovirus/imunologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Animais , Autoantígenos/imunologia , Citotoxicidade Imunológica , Evolução Molecular , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Camundongos , Subfamília A de Receptores Semelhantes a Lectina de Células NK/genética
10.
PLoS Pathog ; 11(2): e1004640, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25659098

RESUMO

Herpesviruses form different gH/gL virion envelope glycoprotein complexes that serve as entry complexes for mediating viral cell-type tropism in vitro; their roles in vivo, however, remained speculative and can be addressed experimentally only in animal models. For murine cytomegalovirus two alternative gH/gL complexes, gH/gL/gO and gH/gL/MCK-2, have been identified. A limitation of studies on viral tropism in vivo has been the difficulty in distinguishing between infection initiation by viral entry into first-hit target cells and subsequent cell-to-cell spread within tissues. As a new strategy to dissect these two events, we used a gO-transcomplemented ΔgO mutant for providing the gH/gL/gO complex selectively for the initial entry step, while progeny virions lack gO in subsequent rounds of infection. Whereas gH/gL/gO proved to be critical for establishing infection by efficient entry into diverse cell types, including liver macrophages, endothelial cells, and hepatocytes, it was dispensable for intra-tissue spread. Notably, the salivary glands, the source of virus for host-to-host transmission, represent an exception in that entry into virus-producing cells did not strictly depend on either the gH/gL/gO or the gH/gL/MCK-2 complex. Only if both complexes were absent in gO and MCK-2 double-knockout virus, in vivo infection was abolished at all sites.


Assuntos
Infecções por Citomegalovirus/transmissão , Citomegalovirus/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Tropismo Viral/fisiologia , Animais , Infecções por Citomegalovirus/metabolismo , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos BALB C
11.
Proc Natl Acad Sci U S A ; 110(41): 16550-5, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24052528

RESUMO

Due to a unique pattern of CD8 T-cell response induced by cytomegaloviruses (CMVs), live attenuated CMVs are attractive candidates for vaccine vectors for a number of clinically relevant infections and tumors. NKG2D is one of the most important activating NK cell receptors that plays a role in costimulation of CD8 T cells. Here we demonstrate that the expression of CD8 T-cell epitope of Listeria monocytogenes by a recombinant mouse CMV (MCMV) expressing the NKG2D ligand retinoic acid early-inducible protein 1-gamma (RAE-1γ) dramatically enhanced the effectiveness and longevity of epitope-specific CD8 T-cell response and conferred protection against a subsequent challenge infection with Listeria monocytogenes. Unexpectedly, the attenuated growth in vivo of the CMV vector expressing RAE-1γ and its capacity to enhance specific CD8 T-cell response were preserved even in mice lacking NKG2D, implying additional immune function for RAE-1γ beyond engagement of NKG2D. Thus, vectors expressing RAE-1γ represent a promising approach in the development of CD8 T-cell-based vaccines.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/imunologia , Vetores Genéticos/imunologia , Evasão da Resposta Imune/imunologia , Proteínas de Membrana/metabolismo , Vacinas Sintéticas/imunologia , Animais , Citomegalovirus/genética , Citometria de Fluxo , Vetores Genéticos/genética , Listeria monocytogenes/imunologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Estatísticas não Paramétricas
12.
Cell Host Microbe ; 13(5): 535-545, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23684305

RESUMO

The immune response against a variety of pathogens can lead to activation of blood formation at ectopic sites, a process termed extramedullary hematopoiesis (EMH). The underlying mechanisms of EMH have been enigmatic. Investigating splenic EMH in mice infected with murine cytomegalovirus (MCMV), we find that, while cells of the adaptive immune system were dispensable for EMH, natural killer (NK) cells were essential. EMH required recognition of infected cells via activating NK cell receptors Ly49H or NKG2D, and correspondingly, viral interference with NK cell recognition abolished EMH. Surprisingly, development of EMH was not induced by NK cell-derived cytokines but was dependent on perforin-mediated cytotoxicity in order to control virus spread. Spreading virus reduced the numbers of F4/80(+) macrophages that were crucial for inflammatory EMH. Hence, whereas MCMV suppresses inflammation-induced EMH, NK cells confine virus spread, thereby protecting extramedullary hematopoietic niches and facilitating EMH.


Assuntos
Hematopoese Extramedular , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Muromegalovirus/patogenicidade , Animais , Modelos Animais de Doenças , Infecções por Herpesviridae/virologia , Células Matadoras Naturais/virologia , Camundongos , Baço/imunologia , Baço/patologia , Baço/virologia
13.
PLoS Pathog ; 8(2): e1002510, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22346748

RESUMO

Cytomegaloviruses express large amounts of viral miRNAs during lytic infection, yet, they only modestly alter the cellular miRNA profile. The most prominent alteration upon lytic murine cytomegalovirus (MCMV) infection is the rapid degradation of the cellular miR-27a and miR-27b. Here, we report that this regulation is mediated by the ∼1.7 kb spliced and highly abundant MCMV m169 transcript. Specificity to miR-27a/b is mediated by a single, apparently optimized, miRNA binding site located in its 3'-UTR. This site is easily and efficiently retargeted to other cellular and viral miRNAs by target site replacement. Expression of the 3'-UTR of m169 by an adenoviral vector was sufficient to mediate its function, indicating that no other viral factors are essential in this process. Degradation of miR-27a/b was found to be accompanied by 3'-tailing and -trimming. Despite its dramatic effect on miRNA stability, we found this interaction to be mutual, indicating potential regulation of m169 by miR-27a/b. Most interestingly, three mutant viruses no longer able to target miR-27a/b, either due to miRNA target site disruption or target site replacement, showed significant attenuation in multiple organs as early as 4 days post infection, indicating that degradation of miR-27a/b is important for efficient MCMV replication in vivo.


Assuntos
Regiões 3' não Traduzidas/genética , Infecções por Citomegalovirus/virologia , MicroRNAs/metabolismo , Muromegalovirus/fisiologia , RNA Viral/metabolismo , Replicação Viral/genética , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sítios de Ligação , Linhagem Celular , Regulação para Baixo/genética , Regulação da Expressão Gênica/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , MicroRNAs/genética , Muromegalovirus/genética , Mutação , Processamento Pós-Transcricional do RNA , Estabilidade de RNA/genética , RNA Viral/genética , Análise de Sequência de RNA
14.
J Exp Med ; 207(12): 2663-73, 2010 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-21078887

RESUMO

Cytomegaloviruses (CMVs) are renowned for interfering with the immune system of their hosts. To sidestep antigen presentation and destruction by CD8(+) T cells, these viruses reduce expression of major histocompatibility complex class I (MHC I) molecules. However, this process sensitizes the virus-infected cells to natural killer (NK) cell-mediated killing via the "missing self" axis. Mouse cytomegalovirus (MCMV) uses m152 and m06 encoded proteins to inhibit surface expression of MHC I molecules. In addition, it encodes another protein, m04, which forms complexes with MHC I and escorts them to the cell surface. This mechanism is believed to prevent NK cell activation and killing by restoring the "self" signature and allowing the engagement of inhibitory Ly49 receptors on NK cells. Here we show that MCMV lacking m04 was attenuated in an NK cell- and MHC I-dependent manner. NK cell-mediated control of the infection was dependent on the presence of NK cell subsets expressing different inhibitory Ly49 receptors. In addition to providing evidence for immunoevasion strategies used by CMVs to avoid NK cell control via the missing-self pathway, our study is the first to demonstrate that missing self-dependent NK cell activation is biologically relevant in the protection against viral infection in vivo.


Assuntos
Proteínas de Transporte/imunologia , Glicoproteínas/imunologia , Evasão da Resposta Imune , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Proteínas Virais/imunologia , Animais , Citotoxicidade Imunológica , Antígenos H-2/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/fisiologia , Proteínas Virais/fisiologia
15.
Curr Opin Microbiol ; 13(4): 530-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20558100

RESUMO

Since their discovery, our understanding of NK cells has evolved from branding them marginal innate immunity cells to key players in anti-viral and anti-tumor immunity. Importance of NK cells in control of various viral infections is perhaps best illustrated by the existence of plethora of viral mechanisms aimed to modulate their function. These mechanisms include not only virally encoded immunoevasion proteins but also viral miRNA. Moreover, the evidence has been accumulated supporting the role of viral immunoevasion of NK cells in viral pathogenesis in vivo.


Assuntos
Células Matadoras Naturais/imunologia , Viroses/virologia , Vírus/imunologia , Animais , Humanos , Evasão da Resposta Imune , Células Matadoras Naturais/virologia , MicroRNAs/imunologia , MicroRNAs/metabolismo , Proteínas/imunologia , Proteínas/metabolismo , RNA Viral/imunologia , RNA Viral/metabolismo , Receptores de Células Matadoras Naturais/imunologia , Receptores de Células Matadoras Naturais/metabolismo , Viroses/imunologia
16.
Immunity ; 31(2): 270-82, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19631564

RESUMO

NKG2D is a potent activating receptor on natural killer (NK) cells and acts as a molecular sensor for stressed cells expressing NKG2D ligands such as infected or tumor-transformed cells. Although NKG2D is expressed on NK cell precursors, its role in NK cell development is not known. We have generated NKG2D-deficient mice by targeting the Klrk1 locus. Here we provide evidence for an important regulatory role of NKG2D in the development of NK cells. The absence of NKG2D caused faster division of NK cells, perturbation in size of some NK cell subpopulations, and their augmented sensitivity to apoptosis. As expected, Klrk1(-/-) NK cells are less responsive to tumor targets expressing NKG2D ligands. Klrk1(-/-) mice, however, showed an enhanced NK cell-mediated resistance to mouse cytomegalovirus infection as a consequence of NK cell dysregulation. Altogether, these findings provide evidence for regulatory function of NKG2D in NK cell physiology.


Assuntos
Infecções por Herpesviridae/veterinária , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Animais , Infecções por Herpesviridae/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Células Matadoras Naturais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Baço/imunologia , Baço/patologia
17.
Nat Immunol ; 3(6): 529-35, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021778

RESUMO

The susceptibility of certain inbred mouse strains to murine cytomegalovirus (MCMV) is related to their inability to generate a strong natural killer (NK) cell response. We addressed here whether the MCMV susceptibility of the BALB/c strain is due to viral functions that control NK cell activation in a strain-specific manner. MCMV expresses two proteins, gp48 and gp40, that are encoded by the genes m06 and m152, respectively; they down-regulate major histocompatibility complex (MHC) class I expression at the plasma membrane. Using MCMV deletion mutants and revertants, we found that gp40 but not gp48 controls NK cell activation. Absence of gp40 improved antiviral NK cell control in BALB/c, but not C57BL/6, mice. Down-regulation of H-60, the high-affinity ligand for the NKG2D receptor, was the mechanism by which gp40 modulates NK cell activation. Thus, a single herpesvirus protein has a dual function in inhibiting both the adaptive as well as the innate immune response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/imunologia , Células Matadoras Naturais/imunologia , Proteínas Virais/imunologia , Animais , Animais Congênicos , Citomegalovirus/genética , Citomegalovirus/patogenicidade , Citotoxicidade Imunológica , Feminino , Deleção de Genes , Genes Virais , Antígenos de Histocompatibilidade Classe I/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores Imunológicos/metabolismo , Receptores de Células Matadoras Naturais , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA