Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Nat Immunol ; 25(2): 240-255, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38182668

RESUMO

Ikaros transcription factors are essential for adaptive lymphocyte function, yet their role in innate lymphopoiesis is unknown. Using conditional genetic inactivation, we show that Ikzf1/Ikaros is essential for normal natural killer (NK) cell lymphopoiesis and IKZF1 directly represses Cish, a negative regulator of interleukin-15 receptor resulting in impaired interleukin-15 receptor signaling. Both Bcl2l11 and BIM levels, and intrinsic apoptosis were increased in Ikzf1-null NK cells, which in part accounts for NK lymphopenia as both were restored to normal levels when Ikzf1 and Bcl2l11 were co-deleted. Ikzf1-null NK cells presented extensive transcriptional alterations with reduced AP-1 transcriptional complex expression and increased expression of Ikzf2/Helios and Ikzf3/Aiolos. IKZF1 and IKZF3 directly bound AP-1 family members and deletion of both Ikzf1 and Ikzf3 in NK cells resulted in further reductions in Jun/Fos expression and complete loss of peripheral NK cells. Collectively, we show that Ikaros family members are important regulators of apoptosis, cytokine responsiveness and AP-1 transcriptional activity.


Assuntos
Células Matadoras Naturais , Fator de Transcrição AP-1 , Fator de Transcrição AP-1/genética , Células Matadoras Naturais/metabolismo , Receptores de Interleucina-15 , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo
2.
Immunity ; 56(3): 531-546.e6, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36773607

RESUMO

Tissue health is dictated by the capacity to respond to perturbations and then return to homeostasis. Mechanisms that initiate, maintain, and regulate immune responses in tissues are therefore essential. Adaptive immunity plays a key role in these responses, with memory and tissue residency being cardinal features. A corresponding role for innate cells is unknown. Here, we have identified a population of innate lymphocytes that we term tissue-resident memory-like natural killer (NKRM) cells. In response to murine cytomegalovirus infection, we show that circulating NK cells were recruited in a CX3CR1-dependent manner to the salivary glands where they formed NKRM cells, a long-lived, tissue-resident population that prevented autoimmunity via TRAIL-dependent elimination of CD4+ T cells. Thus, NK cells develop adaptive-like features, including long-term residency in non-lymphoid tissues, to modulate inflammation, restore immune equilibrium, and preserve tissue health. Modulating the functions of NKRM cells may provide additional strategies to treat inflammatory and autoimmune diseases.


Assuntos
Infecções por Citomegalovirus , Muromegalovirus , Humanos , Animais , Camundongos , Células Matadoras Naturais , Imunidade Adaptativa , Linfócitos T , Imunidade Inata
4.
Nat Immunol ; 17(7): 816-24, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27213690

RESUMO

The detection of aberrant cells by natural killer (NK) cells is controlled by the integration of signals from activating and inhibitory ligands and from cytokines such as IL-15. We identified cytokine-inducible SH2-containing protein (CIS, encoded by Cish) as a critical negative regulator of IL-15 signaling in NK cells. Cish was rapidly induced in response to IL-15, and deletion of Cish rendered NK cells hypersensitive to IL-15, as evidenced by enhanced proliferation, survival, IFN-γ production and cytotoxicity toward tumors. This was associated with increased JAK-STAT signaling in NK cells in which Cish was deleted. Correspondingly, CIS interacted with the tyrosine kinase JAK1, inhibiting its enzymatic activity and targeting JAK for proteasomal degradation. Cish(-/-) mice were resistant to melanoma, prostate and breast cancer metastasis in vivo, and this was intrinsic to NK cell activity. Our data uncover a potent intracellular checkpoint in NK cell-mediated tumor immunity and suggest possibilities for new cancer immunotherapies directed at blocking CIS function.


Assuntos
Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Neoplasias/terapia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Proliferação de Células/genética , Citotoxicidade Imunológica/genética , Vigilância Imunológica , Interferon gama/metabolismo , Interleucina-15/metabolismo , Janus Quinase 1/metabolismo , Ativação Linfocitária/genética , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Terapia de Alvo Molecular , Neoplasias/imunologia , Transdução de Sinais/genética , Proteínas Supressoras da Sinalização de Citocina/genética
5.
Immunity ; 50(6): 1337-1339, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216454

RESUMO

Natural killer (NK) cells display some features equivalent to those of adaptive immune effectors, but the molecular processes underlying these adaptive-like characteristics are just beginning to be defined. In this issue of Immunity, Adams et al. (2019) and Grassmann et al. (2019) report that avidity selection for Ly49H governs the expansion, differentiation, and function of NK cells after cytomegalovirus infection.


Assuntos
Infecções por Citomegalovirus , Diferenciação Celular , Humanos , Células Matadoras Naturais
6.
Immunol Rev ; 323(1): 54-60, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38568046

RESUMO

Natural killer (NK) cells are the prototype innate effector lymphocyte population that plays an important role in controlling viral infections and tumors. Studies demonstrating that NK cells form long-lived memory populations, akin to those generated by adaptive immune cells, prompted a revaluation of the potential functions of NK cells. Recent data demonstrating that NK cells are recruited from the circulation into tissues where they form long-lived memory-like populations further emphasize that NK cells have properties that mirror those of adaptive immune cells. NK cells that localize in non-lymphoid tissues are heterogeneous, and there is a growing appreciation that immune responses occurring within tissues are subject to tissue-specific regulation. Here we discuss both the immune effector and immunoregulatory functions of NK cells, with a particular emphasis on the role of NK cells within non-lymphoid tissues and how the tissue microenvironment shapes NK cell-dependent outcomes.


Assuntos
Memória Imunológica , Células Matadoras Naturais , Células Matadoras Naturais/imunologia , Humanos , Animais , Especificidade de Órgãos/imunologia , Imunidade Adaptativa , Microambiente Celular , Imunidade Inata
8.
J Virol ; 96(12): e0041922, 2022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35638820

RESUMO

Myxovirus resistance (Mx) proteins are dynamin-like GTPases that are inducible by interferons (IFNs) following virus infections. Most studies investigating Mx proteins have focused on their activity against influenza A viruses (IAV), although emerging evidence suggests that some Mx proteins may exhibit broader antiviral activity. Herein, we demonstrate that in addition to IAV, overexpression of mouse Mx1 (mMx1), but not mMx2, resulted in potent inhibition of growth of the human alphaherpesviruses herpes simplex virus 1 (HSV-1) and HSV-2, whereas neither inhibited the mouse betaherpesvirus murine cytomegalovirus (MCMV) in vitro. IFN induction of a functional endogenous mMx1 in primary mouse fibroblasts ex vivo was also associated with inhibition of HSV-1 growth. Using an in vitro overexpression approach, we demonstrate that mutations that result in redistribution of mMx1 from the nucleus to the cytoplasm or in loss of its combined GTP binding and GTPase activity also abrogated its ability to inhibit HSV-1 growth. Overexpressed mMx1 did not inhibit early HSV-1 gene expression but was shown to inhibit both replication of the HSV-1 genome as well as subsequent late gene expression. In a mouse model of cutaneous HSV-1 infection, mice expressing a functional endogenous mMx1 showed significant reductions in the severity of skin lesions as well as reduced HSV-1 titers in both the skin and dorsal root ganglia (DRG). Together, these data demonstrate that mMx1 mediates potent antiviral activity against human alphaherpesviruses by blocking replication of the viral genome and subsequent steps in virus replication. Moreover, endogenous mMx1 potently inhibited pathogenesis in the zosteriform mouse model of HSV-1 infection. IMPORTANCE While a number of studies have demonstrated that human Mx proteins can inhibit particular herpesviruses in vitro, we are the first to report the antiviral activity of mouse Mx1 (mMx1) against alphaherpesviruses both in vitro and in vivo. We demonstrate that both overexpressed mMx1 and endogenous mMx1 potently restrict HSV-1 growth in vitro. mMx1-mediated inhibition of HSV-1 was not associated with inhibition of virus entry and/or import of the viral genome into the nucleus, but rather with inhibition of HSV-1 genomic replication as well as subsequent late gene expression. Therefore, inhibition of human alphaherpesviruses by mMx1 occurs by a mechanism that is distinct from that reported for human Mx proteins against herpesviruses. Importantly, we also provide evidence that expression of a functional endogenous mMx1 can limit HSV-1 pathogenesis in a mouse model of infection.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Proteínas de Resistência a Myxovirus , Replicação Viral , Animais , Modelos Animais de Doenças , Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Interferons/metabolismo , Camundongos , Muromegalovirus , Proteínas de Resistência a Myxovirus/metabolismo
9.
Immunity ; 41(4): 646-56, 2014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-25367576

RESUMO

Natural killer (NK) cells have been reported to control adaptive immune responses that occur in lymphoid organs at the early stages of immune challenge. The physiological purpose of such regulatory activity remains unclear, because it generally does not confer a survival advantage. We found that NK cells specifically eliminated activated CD4(+) T cells in the salivary gland during chronic murine cytomegalovirus (MCMV) infection. This was dependent on TNF-related apoptosis inducing ligand (TRAIL) expression by NK cells. Although NK cell-mediated deletion of CD4(+) T cells prolonged the chronicity of infection, it also constrained viral-induced autoimmunity. In the absence of this activity, chronic infection was associated with a Sjogren's-like syndrome characterized by focal lymphocytic infiltration into the glands, production of autoantibodies, and reduced saliva and tear secretion. Thus, NK cells are an important homeostatic control that balances the efficacy of adaptive immune responses with the risk of developing autoimmunity.


Assuntos
Autoimunidade/imunologia , Linfócitos T CD4-Positivos/imunologia , Infecções por Herpesviridae/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Transferência Adotiva , Animais , Apoptose/imunologia , Doença Crônica , Citotoxicidade Imunológica/imunologia , Células Matadoras Naturais/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK/biossíntese , Glândulas Salivares/imunologia , Glândulas Salivares/patologia , Glândulas Salivares/virologia
10.
PLoS Genet ; 16(3): e1008604, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32130224

RESUMO

The influence of environmental insults on the onset and progression of mitochondrial diseases is unknown. To evaluate the effects of infection on mitochondrial disease we used a mouse model of Leigh Syndrome, where a missense mutation in the Taco1 gene results in the loss of the translation activator of cytochrome c oxidase subunit I (TACO1) protein. The mutation leads to an isolated complex IV deficiency that mimics the disease pathology observed in human patients with TACO1 mutations. We infected Taco1 mutant and wild-type mice with a murine cytomegalovirus and show that a common viral infection exacerbates the complex IV deficiency in a tissue-specific manner. We identified changes in neuromuscular morphology and tissue-specific regulation of the mammalian target of rapamycin pathway in response to viral infection. Taken together, we report for the first time that a common stress condition, such as viral infection, can exacerbate mitochondrial dysfunction in a genetic model of mitochondrial disease.


Assuntos
Deficiência de Citocromo-c Oxidase/genética , Infecções por Citomegalovirus/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Muromegalovirus/patogenicidade , Animais , Deficiência de Citocromo-c Oxidase/virologia , Infecções por Citomegalovirus/virologia , Modelos Animais de Doenças , Doença de Leigh/genética , Doença de Leigh/virologia , Camundongos , Camundongos Endogâmicos C57BL , Doenças Mitocondriais/virologia , Mutação/genética , Serina-Treonina Quinases TOR/genética
11.
Immunol Cell Biol ; 98(10): 802-804, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33016375

RESUMO

Ng et al. have identified NKG7 as a regulator of inflammation in response to diverse immunological challenges. While NKG7 was required for the degranulation of cytotoxic cells, additional defects including reduced expansion and trafficking of CD8 T cells, and altered antigen presentation, were noted in NKG7-deficient mice. The precise mechanism by which NKG7 mediates its effects has not been resolved but may involve regulation of endosomal vesicle trafficking.


Assuntos
Endossomos , Células Matadoras Naturais , Animais , Linfócitos T CD8-Positivos , Células Cultivadas , Exocitose , Inflamação , Proteínas de Membrana , Camundongos
12.
PLoS Pathog ; 14(5): e1007040, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29852019

RESUMO

Recent outbreaks of Ebola and Zika have highlighted the possibility that viruses may cause enduring infections in tissues like the eye, including the neural retina, which have been considered immune privileged. Whether this is a peculiarity of exotic viruses remains unclear, since the impact of more common viral infections on neural compartments has not been examined, especially in immunocompetent hosts. Cytomegalovirus is a common, universally distributed pathogen, generally innocuous in healthy individuals. Whether in immunocompetent hosts cytomegalovirus can access the eye, and reside there indefinitely, was unknown. Using the well-established murine cytomegalovirus infection model, we show that systemic infection of immunocompetent hosts results in broad ocular infection, chronic inflammation and establishment of a latent viral pool in the eye. Infection leads to infiltration and accumulation of anti-viral CD8+ T cells in the eye, and to the development of tissue resident memory T cells that localize to the eye, including the retina. These findings identify the eye as an unexpected reservoir for cytomegalovirus, and suggest that common viruses may target this organ more frequently than appreciated. Notably, they also highlight that infection triggers sustained inflammatory responses in the eye, including the neural retina.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Olho/virologia , Animais , Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Modelos Animais de Doenças , Reservatórios de Doenças/microbiologia , Olho/imunologia , Feminino , Memória Imunológica/imunologia , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/fisiologia , Linfócitos T/imunologia , Viroses
13.
EMBO J ; 33(22): 2721-34, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25319415

RESUMO

Natural killer (NK) cells are an innate lymphoid cell lineage characterized by their capacity to provide rapid effector functions, including cytokine production and cytotoxicity. Here, we identify the Ikaros family member, Aiolos, as a regulator of NK-cell maturation. Aiolos expression is initiated at the point of lineage commitment and maintained throughout NK-cell ontogeny. Analysis of cell surface markers representative of distinct stages of peripheral NK-cell maturation revealed that Aiolos was required for the maturation in the spleen of CD11b(high)CD27(-) NK cells. The differentiation block was intrinsic to the NK-cell lineage and resembled that found in mice lacking either T-bet or Blimp1; however, genetic analysis revealed that Aiolos acted independently of all other known regulators of NK-cell differentiation. NK cells lacking Aiolos were strongly hyper-reactive to a variety of NK-cell-mediated tumor models, yet impaired in controlling viral infection, suggesting a regulatory function for CD27(-) NK cells in balancing these two arms of the immune response. These data place Aiolos in the emerging gene regulatory network controlling NK-cell maturation and function.


Assuntos
Diferenciação Celular/imunologia , Imunidade Celular , Células Matadoras Naturais/imunologia , Transativadores/imunologia , Animais , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Diferenciação Celular/genética , Redes Reguladoras de Genes/imunologia , Fator de Transcrição Ikaros , Células Matadoras Naturais/citologia , Camundongos , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Viroses/genética , Viroses/imunologia
14.
Proc Natl Acad Sci U S A ; 112(18): E2376-84, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25901316

RESUMO

Natural killer (NK) cells are naturally circulating innate lymphoid cells that protect against tumor initiation and metastasis and contribute to immunopathology during inflammation. The signals that prime NK cells are not completely understood, and, although the importance of IFN type I is well recognized, the role of type III IFN is comparatively very poorly studied. IL-28R-deficient mice were resistant to LPS and cecal ligation puncture-induced septic shock, and hallmark cytokines in these disease models were dysregulated in the absence of IL-28R. IL-28R-deficient mice were more sensitive to experimental tumor metastasis and carcinogen-induced tumor formation than WT mice, and additional blockade of interferon alpha/beta receptor 1 (IFNAR1), but not IFN-γ, further enhanced metastasis and tumor development. IL-28R-deficient mice were also more susceptible to growth of the NK cell-sensitive lymphoma, RMAs. Specific loss of IL-28R in NK cells transferred into lymphocyte-deficient mice resulted in reduced LPS-induced IFN-γ levels and enhanced tumor metastasis. Therefore, by using IL-28R-deficient mice, which are unable to signal type III IFN-λ, we demonstrate for the first time, to our knowledge, the ability of IFN-λ to directly regulate NK cell effector functions in vivo, alone and in the context of IFN-αß.


Assuntos
Interferon Tipo I/metabolismo , Células Matadoras Naturais/citologia , Receptores de Citocinas/metabolismo , Animais , Carcinógenos , Separação Celular , Citocinas/metabolismo , Endotoxinas/metabolismo , Citometria de Fluxo , Deleção de Genes , Regulação da Expressão Gênica , Lipopolissacarídeos/metabolismo , Masculino , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Metástase Neoplásica , RNA Mensageiro/metabolismo , Choque Séptico/metabolismo , Transdução de Sinais
15.
Blood ; 126(12): 1503-14, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26130706

RESUMO

Viral infection is a common, life-threatening complication after allogeneic bone marrow transplantation (BMT), particularly in the presence of graft-versus-host disease (GVHD). Using cytomegalovirus (CMV) as the prototypic pathogen, we have delineated the mechanisms responsible for the inability to mount protective antiviral responses in this setting. Although CMV infection was self-limiting after syngeneic BMT, in the presence of GVHD after allogeneic BMT, CMV induced a striking cytopathy resulting in universal mortality in conjunction with a fulminant necrotizing hepatitis. Critically, GVHD induced a profound dendritic cell (DC) defect that led to a failure in the generation of CMV-specific CD8(+) T-cell responses. This was accompanied by a defect in antiviral CD8(+) T cells. In combination, these defects dramatically limited antiviral T-cell responses. The transfer of virus-specific cells circumvented the DC defects and provided protective immunity, despite concurrent GVHD. These data demonstrate the importance of avoiding GVHD when reconstructing antiviral immunity after BMT, and highlight the mechanisms by which the adoptive transfer of virus-specific T cells overcome the endogenous defects in priming invoked by GVHD.


Assuntos
Linfócitos T CD8-Positivos/patologia , Infecções por Citomegalovirus/etiologia , Citomegalovirus/imunologia , Células Dendríticas/patologia , Doença Enxerto-Hospedeiro/complicações , Transferência Adotiva , Animais , Transplante de Medula Óssea/efeitos adversos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Células Cultivadas , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/terapia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Feminino , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Doença Enxerto-Hospedeiro/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
16.
J Immunol ; 195(3): 1233-41, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26116505

RESUMO

Inflammasomes are protein complexes that promote caspase activation, resulting in processing of IL-1ß and cell death, in response to infection and cellular stresses. Inflammasomes have been anticipated to contribute to autoimmunity. The New Zealand Black (NZB) mouse develops anti-erythrocyte Abs and is a model of autoimmune hemolytic anemia. These mice also develop anti-nuclear Abs typical of lupus. In this article, we show that NZB macrophages have deficient inflammasome responses to a DNA virus and fungal infection. Absent in melanoma 2 (AIM2) inflammasome responses are compromised in NZB by high expression of the AIM 2 antagonist protein p202, and consequently NZB cells had low IL-1ß output in response to both transfected DNA and mouse CMV infection. Surprisingly, we also found that a second inflammasome system, mediated by the NLR family, pyrin domain containing 3 (NLRP3) initiating protein, was completely lacking in NZB cells. This was due to a point mutation in an intron of the Nlrp3 gene in NZB mice, which generates a novel splice acceptor site. This leads to incorporation of a pseudoexon with a premature stop codon. The lack of full-length NLRP3 protein results in NZB being effectively null for Nlrp3, with no production of bioactive IL-1ß in response to NLRP3 stimuli, including infection with Candida albicans. Thus, this autoimmune strain harbors two inflammasome deficiencies, mediated through quite distinct mechanisms. We hypothesize that the inflammasome deficiencies in NZB alter the interaction of the host with both microflora and pathogens, promoting prolonged production of cytokines that contribute to development of autoantibodies.


Assuntos
Anemia Hemolítica Autoimune/genética , Proteínas de Transporte/genética , Proteínas de Ligação a DNA/genética , Inflamassomos/genética , Macrófagos/imunologia , Anemia Hemolítica Autoimune/imunologia , Animais , Anticorpos Antinucleares/imunologia , Autoimunidade/genética , Autoimunidade/imunologia , Candida albicans/imunologia , Candidíase/imunologia , Candidíase/microbiologia , Proteínas de Transporte/imunologia , Caspase 1/genética , Citomegalovirus/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/imunologia , Inflamassomos/imunologia , Interleucina-1beta/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Camundongos , Camundongos Endogâmicos NZB , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais/genética , Transdução de Sinais/imunologia
17.
PLoS Pathog ; 10(12): e1004526, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25502180

RESUMO

Many immune response genes are highly polymorphic, consistent with the selective pressure imposed by pathogens over evolutionary time, and the need to balance infection control with the risk of auto-immunity. Epidemiological and genomic studies have identified many genetic variants that confer susceptibility or resistance to pathogenic micro-organisms. While extensive polymorphism has been reported for the granzyme B (GzmB) gene, its relevance to pathogen immunity is unexplored. Here, we describe the biochemical and cytotoxic functions of a common allele of GzmB (GzmBW) common in wild mouse. While retaining 'Asp-ase' activity, GzmBW has substrate preferences that differ considerably from GzmBP, which is common to all inbred strains. In vitro, GzmBW preferentially cleaves recombinant Bid, whereas GzmBP activates pro-caspases directly. Recombinant GzmBW and GzmBP induced equivalent apoptosis of uninfected targets cells when delivered with perforin in vitro. Nonetheless, mice homozygous for GzmBW were unable to control murine cytomegalovirus (MCMV) infection, and succumbed as a result of excessive liver damage. Although similar numbers of anti-viral CD8 T cells were generated in both mouse strains, GzmBW-expressing CD8 T cells isolated from infected mice were unable to kill MCMV-infected targets in vitro. Our results suggest that known virally-encoded inhibitors of the intrinsic (mitochondrial) apoptotic pathway account for the increased susceptibility of GzmBW mice to MCMV. We conclude that different natural variants of GzmB have a profound impact on the immune response to a common and authentic viral pathogen.


Assuntos
Variação Genética/genética , Granzimas/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/mortalidade , Muromegalovirus/imunologia , Viroses/imunologia , Viroses/mortalidade , Alelos , Sequência de Aminoácidos , Animais , Apoptose , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Caspases/metabolismo , Modelos Animais de Doenças , Granzimas/análise , Granzimas/deficiência , Infecções por Herpesviridae/patologia , Imunidade Inata/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Viroses/patologia
18.
Eur J Immunol ; 44(2): 409-19, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24166710

RESUMO

Cytomegalovirus latently infects myeloid cells; however, the acute effects of the virus on this cell subset are poorly characterised. We demonstrate that systemic cytomegalovirus infection induced rapid activation of monocytes in the bone marrow, characterised by upregulation of CD69, CD11c, Ly6C and M-CSF receptor. Activated bone marrow monocytes were more sensitive to M-CSF and less sensitive to granulocyte-monocyte colony stimulating factor in vitro, resulting in the generation of more macrophages and fewer dendritic cells, respectively. Monocyte activation was also observed in the periphery and resulted in significant accumulation of monocytes in the spleen. MyD88 expression was required within the haematopoietic compartment to initiate monocyte activation and recruitment. However, monocytes lacking MyD88 were activated and recruited in the presence of MyD88-sufficient cells in mixed bone marrow chimeras, indicating that once initiated, the process was MyD88 independent. Interestingly, we found that monocyte activation occurred in the absence of the common inflammatory cytokines, namely type I interferons (IFNs), IL-6, TNF-α and IL-1 as well as the NLRP3 inflammasome adaptor protein, ASC. We also excluded a role for the chemokine-like protein MCK-2 (m131/129) expressed by murine CMV. Taken together, these results challenge the notion that a single inflammatory cytokine mediates activation and recruitment of monocytes in response to infection.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos Ly/imunologia , Antígenos Ly/metabolismo , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/virologia , Antígeno CD11c/imunologia , Antígeno CD11c/metabolismo , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Células Cultivadas , Quimiocinas CC/imunologia , Quimiocinas CC/metabolismo , Infecções por Citomegalovirus/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Feminino , Inflamação/virologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Interleucina-1/imunologia , Interleucina-1/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Fator Estimulador de Colônias de Macrófagos/imunologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/virologia , Fator 88 de Diferenciação Mieloide/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Baço/imunologia , Baço/metabolismo , Baço/virologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
19.
PLoS Pathog ; 9(2): e1003192, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23468630

RESUMO

Successful replication and transmission of large DNA viruses such as the cytomegaloviruses (CMV) family of viruses depends on the ability to interfere with multiple aspects of the host immune response. Apoptosis functions as a host innate defence mechanism against viral infection, and the capacity to interfere with this process is essential for the replication of many viruses. The Bcl-2 family of proteins are the principle regulators of apoptosis, with two pro-apoptotic members, Bax and Bak, essential for apoptosis to proceed. The m38.5 protein encoded by murine CMV (MCMV) has been identified as Bax-specific inhibitor of apoptosis. Recently, m41.1, a protein product encoded by the m41 open reading frame (ORF) of MCMV, has been shown to inhibit Bak activity in vitro. Here we show that m41.1 is critical for optimal MCMV replication in vivo. Growth of a m41.1 mutant was attenuated in multiple organs, a defect that was not apparent in Bak(-/-) mice. Thus, m41.1 promotes MCMV replication by inhibiting Bak-dependent apoptosis during in vivo infection. The results show that Bax and Bak mediate non-redundant functions during MCMV infection and that the virus produces distinct inhibitors for each protein to counter the activity of these proteins.


Assuntos
Apoptose/fisiologia , Proteínas Inibidoras de Apoptose/metabolismo , Muromegalovirus/patogenicidade , Replicação Viral/fisiologia , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Imunidade Inata , Proteínas Inibidoras de Apoptose/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Muromegalovirus/metabolismo , Proteína X Associada a bcl-2/metabolismo
20.
Blood ; 122(1): 55-60, 2013 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-23673858

RESUMO

Major histocompatibility complex class I-restricted T-cell immunity is essential to control infection with cytomegalovirus (CMV), a clinically important virus that causes significant disease in immunocompromised individuals. Cross-presentation is considered the primary mode of antigen presentation to generate protective antiviral CD8⁺ T-cell immunity. Herpesviruses, including CMV, encode numerous proteins that interfere with direct antigen presentation, leading to the paradigm that T-cell immunity to these pathogens necessitates cross-presentation. However, the antigen presentation requirements needed to generate a protective T-cell response to CMV remain unknown. Here, we show that a fully functional antiviral CD8⁺ T-cell response can be generated in a system where cross-presentation is shut down by pretreatment with CpG. Notably, in this setting, CD8⁺ T cells demonstrate accelerated control of infection, and organ pathology is limited. These data indicate that protective antiviral T-cell immunity to CMV is generated by direct presentation and can be enhanced by pretreatment with CpG.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/terapia , Citomegalovirus/imunologia , Oligodesoxirribonucleotídeos/farmacologia , Adjuvantes Imunológicos/farmacologia , Animais , Apresentação de Antígeno/imunologia , Apresentação Cruzada/imunologia , Citomegalovirus/crescimento & desenvolvimento , Hospedeiro Imunocomprometido/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Baço/imunologia , Baço/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA