Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 38: 511-539, 2020 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-32340578

RESUMO

The continuous interactions between host and pathogens during their coevolution have shaped both the immune system and the countermeasures used by pathogens. Natural killer (NK) cells are innate lymphocytes that are considered central players in the antiviral response. Not only do they express a variety of inhibitory and activating receptors to discriminate and eliminate target cells but they can also produce immunoregulatory cytokines to alert the immune system. Reciprocally, several unrelated viruses including cytomegalovirus, human immunodeficiency virus, influenza virus, and dengue virus have evolved a multitude of mechanisms to evade NK cell function, such as the targeting of pathways for NK cell receptors and their ligands, apoptosis, and cytokine-mediated signaling. The studies discussed in this article provide further insights into the antiviral function of NK cells and the pathways involved, their constituent proteins, and ways in which they could be manipulated for host benefit.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Vírus/imunologia , Animais , Biomarcadores , Citocinas/metabolismo , Humanos , Receptores de Células Matadoras Naturais/metabolismo , Transdução de Sinais , Viroses/imunologia , Viroses/metabolismo , Viroses/virologia
2.
Nat Immunol ; 18(1): 54-63, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27721430

RESUMO

Genes and pathways in which inactivation dampens tissue inflammation present new opportunities for understanding the pathogenesis of common human inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis and multiple sclerosis. We identified a mutation in the gene encoding the deubiquitination enzyme USP15 (Usp15L749R) that protected mice against both experimental cerebral malaria (ECM) induced by Plasmodium berghei and experimental autoimmune encephalomyelitis (EAE). Combining immunophenotyping and RNA sequencing in brain (ECM) and spinal cord (EAE) revealed that Usp15L749R-associated resistance to neuroinflammation was linked to dampened type I interferon responses in situ. In hematopoietic cells and in resident brain cells, USP15 was coexpressed with, and functionally acted together with the E3 ubiquitin ligase TRIM25 to positively regulate type I interferon responses and to promote pathogenesis during neuroinflammation. The USP15-TRIM25 dyad might be a potential target for intervention in acute or chronic states of neuroinflammation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Malária Cerebral/imunologia , Inflamação Neurogênica/imunologia , Fatores de Transcrição/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Encefalomielite Autoimune Experimental/tratamento farmacológico , Células HEK293 , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Malária Cerebral/tratamento farmacológico , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Terapia de Alvo Molecular , Glicoproteína Mielina-Oligodendrócito/imunologia , Inflamação Neurogênica/tratamento farmacológico , Fragmentos de Peptídeos/imunologia , Plasmodium berghei/imunologia , Fatores de Transcrição/genética , Proteases Específicas de Ubiquitina/genética
3.
Nat Immunol ; 17(1): 65-75, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26595887

RESUMO

Viral respiratory tract infections are the main causative agents of the onset of infection-induced asthma and asthma exacerbations that remain mechanistically unexplained. Here we found that deficiency in signaling via type I interferon receptor led to deregulated activation of group 2 innate lymphoid cells (ILC2 cells) and infection-associated type 2 immunopathology. Type I interferons directly and negatively regulated mouse and human ILC2 cells in a manner dependent on the transcriptional activator ISGF3 that led to altered cytokine production, cell proliferation and increased cell death. In addition, interferon-γ (IFN-γ) and interleukin 27 (IL-27) altered ILC2 function dependent on the transcription factor STAT1. These results demonstrate that type I and type II interferons, together with IL-27, regulate ILC2 cells to restrict type 2 immunopathology.


Assuntos
Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Linfócitos/imunologia , Infecções Respiratórias/imunologia , Animais , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Infecções Respiratórias/patologia
4.
Proc Natl Acad Sci U S A ; 119(32): e2204539119, 2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35878012

RESUMO

Viruses evade the innate immune response by suppressing the production or activity of cytokines such as type I interferons (IFNs). Here we report the discovery of a mechanism by which the SARS-CoV-2 virus coopts an intrinsic cellular machinery to suppress the production of the key immunostimulatory cytokine IFN-ß. We reveal that the SARS-CoV-2 encoded nonstructural protein 2 (NSP2) directly interacts with the cellular GIGYF2 protein. This interaction enhances the binding of GIGYF2 to the mRNA cap-binding protein 4EHP, thereby repressing the translation of the Ifnb1 mRNA. Depletion of GIGYF2 or 4EHP significantly enhances IFN-ß production, which inhibits SARS-CoV-2 replication. Our findings reveal a target for rescuing the antiviral innate immune response to SARS-CoV-2 and other RNA viruses.


Assuntos
COVID-19 , Proteínas de Transporte , Interferon Tipo I , Proteínas não Estruturais Virais , COVID-19/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/genética , SARS-CoV-2 , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
5.
Nat Immunol ; 13(9): 843-50, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22863752

RESUMO

A large gap in our understanding of infant immunity is why natural killer (NK) cell responses are deficient, which makes infants more prone to viral infection. Here we demonstrate that transforming growth factor-ß (TGF-ß) was responsible for NK cell immaturity during infancy. We found more fully mature NK cells in CD11c(dnR) mice, whose NK cells lack TGF-ß receptor (TGF-ßR) signaling. Ontogenic maturation of NK cells progressed faster in the absence of TGF-ß signaling, which results in the formation of a mature NK cell pool early in life. As a consequence, infant CD11c(dnR) mice efficiently controlled viral infections. These data thus demonstrate an unprecedented role for TGF-ß in ontogeny that can explain why NK cell responses are deficient early in life.


Assuntos
Sistema Imunitário/citologia , Sistema Imunitário/crescimento & desenvolvimento , Células Matadoras Naturais/citologia , Fator de Crescimento Transformador beta/imunologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/imunologia , Citometria de Fluxo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células-Tronco , Fator de Crescimento Transformador beta/metabolismo
6.
Photochem Photobiol Sci ; 23(2): 339-354, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38308169

RESUMO

Ultraviolet radiation's germicidal efficacy depends on several parameters, including wavelength, radiant exposure, microbial physiology, biological matrices, and surfaces. In this work, several ultraviolet radiation sources (a low-pressure mercury lamp, a KrCl excimer, and four UV LEDs) emitting continuous or pulsed irradiation were compared. The greatest log reductions in E. coli cells and B. subtilis endospores were 4.1 ± 0.2 (18 mJ cm-2) and 4.5 ± 0.1 (42 mJ cm-2) with continuous 222 nm, respectively. The highest MS2 log reduction observed was 2.7 ± 0.1 (277 nm at 3809 mJ cm-2). Log reductions of SARS-CoV-2 with continuous 222 nm and 277 nm were ≥ 3.4 ± 0.7, with 13.3 mJ cm-2 and 60 mJ cm-2, respectively. There was no statistical difference between continuous and pulsed irradiation (0.83-16.7% [222 nm and 277 nm] or 0.83-20% [280 nm] duty rates) on E. coli inactivation. Pulsed 260 nm radiation (0.5% duty rate) at 260 nm yielded significantly greater log reduction for both bacteria than continuous 260 nm radiation. There was no statistical difference in SARS-CoV-2 inactivation between continuous and pulsed 222 nm UV-C radiation and pulsed 277 nm radiation demonstrated greater germicidal efficacy than continuous 277 nm radiation. Greater radiant exposure for all radiation sources was required to inactivate MS2 bacteriophage. Findings demonstrate that pulsed irradiation could be more useful than continuous UV radiation in human-occupied spaces, but threshold limit values should be respected. Pathogen-specific sensitivities, experimental setup, and quantification methods for determining germicidal efficacy remain important factors when optimizing ultraviolet radiation for surface decontamination or other applications.


Assuntos
COVID-19 , Raios Ultravioleta , Humanos , SARS-CoV-2 , Escherichia coli/efeitos da radiação , Desinfecção/métodos
7.
Immunology ; 170(4): 553-566, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37688495

RESUMO

Rheumatoid arthritis is a chronic and systemic inflammatory disease that affects approximately 1% of the world's population and is characterised by joint inflammation, the destruction of articular cartilage and bone, and many potentially life-threatening extraarticular manifestations. B lymphocytes play a central role in the pathology of rheumatoid arthritis as the precursors of autoantibody secreting plasma cells, as highly potent antigen-presenting cells, and as a source of various inflammatory cytokines, however, the effects of rheumatoid arthritis on B lymphocyte development remain poorly understood. Here, we analyse B lymphocyte development in murine models of rheumatoid arthritis, quantifying all the subsets of B cell precursors in the bone marrow and splenic B cells using flow cytometry. We demonstrate a severe reduction in pre-B cells and immature B cells in the bone marrow of mice with active disease, despite no major effects on the mature naïve B cell numbers. The loss of B cell precursors in the bone marrow of the affected mice was associated with a highly significant reduction in the proportion of Ki67+ cells, indicating impaired cell proliferation, while the viability of the B cell precursors was not significantly affected. We also observed some mobilisation of the B cell precursor cells into the mouse spleen, demonstrated with flow cytometry and pre-B colony forming units assays. In summary, the current work demonstrates a severe dysregulation in B lymphocyte development in murine rheumatoid arthritis, with possible implications for B cell repertoire formation, tolerance induction, and disease mechanisms.


Assuntos
Artrite Experimental , Artrite Reumatoide , Camundongos , Animais , Modelos Animais de Doenças , Linfócitos B , Tolerância Imunológica
9.
J Immunol ; 202(5): 1479-1493, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30683700

RESUMO

Herpes simplex encephalitis (HSE), caused by HSV type 1 (HSV-1) infection, is an acute neuroinflammatory condition of the CNS and remains the most common type of sporadic viral encephalitis worldwide. Studies in humans have shown that susceptibility to HSE depends in part on the genetic make-up of the host, with deleterious mutations in the TLR3/type I IFN axis underlying some cases of childhood HSE. Using an in vivo chemical mutagenesis screen for HSV-1 susceptibility in mice, we identified a susceptible pedigree carrying a causal truncating mutation in the Rel gene (RelC307X ), encoding for the NF-κB transcription factor subunit c-Rel. Like Myd88-/- and Irf3-/- mice, RelC307X mice were susceptible to intranasal HSV-1 infection. Reciprocal bone marrow transfers into lethally irradiated hosts suggested that defects in both hematopoietic and CNS-resident cellular compartments contributed together to HSE susceptibility in RelC307X mice. Although the RelC307X mutation maintained cell-intrinsic antiviral control, it drove increased apoptotic cell death in infected fibroblasts. Moreover, reduced numbers of CD4+CD25+Foxp3+ T regulatory cells, and dysregulated NK cell and CD4+ effector T cell responses in infected RelC307X animals, indicated that protective immunity was also compromised in these mice. In the CNS, moribund RelC307X mice failed to control HSV-1 viral replication in the brainstem and cerebellum, triggering cell death and elevated expression of Ccl2, Il6, and Mmp8 characteristic of HSE neuroinflammation and pathology. In summary, our work implicates c-Rel in both CNS-resident cell survival and lymphocyte responses to HSV-1 infection and as a novel cause of HSE disease susceptibility in mice.


Assuntos
Sistema Nervoso Central/imunologia , Encefalite por Herpes Simples/imunologia , Inflamação/imunologia , Replicação Viral/imunologia , Animais , Chlorocebus aethiops , Encefalite por Herpes Simples/virologia , Inflamação/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Vero
10.
Infect Immun ; 88(2)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31792077

RESUMO

We used a genome-wide screen in N-ethyl-N-nitrosourea (ENU)-mutagenized mice to identify genes in which recessive loss-of-function mutations protect against pathological neuroinflammation. We identified an R367Q mutation in the ZBTB7B (ThPOK) protein in which homozygosity causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA. Zbtb7bR367Q homozygous mice show a defect in the lymphoid compartment expressed as severe reduction in the number of single-positive CD4 T cells in the thymus and in the periphery, reduced brain infiltration of proinflammatory leukocytes in P. berghei ANKA-infected mice, and reduced production of proinflammatory cytokines by primary T cells ex vivo and in vivo Dampening of proinflammatory immune responses in Zbtb7bR367Q mice is concomitant to increased susceptibility to infection with avirulent (Mycobacterium bovis BCG) and virulent (Mycobacterium tuberculosis H37Rv) mycobacteria. The R367Q mutation maps to the first DNA-binding zinc finger domain of ThPOK and causes loss of base contact by R367 in the major groove of the DNA, which is predicted to impair DNA binding. Global immunoprecipitation of ThPOK-containing chromatin complexes coupled to DNA sequencing (ChIP-seq) identified transcriptional networks and candidate genes likely to play key roles in CD4+ CD8+ T cell development and in the expression of lineage-specific functions of these cells. This study highlights ThPOK as a global regulator of immune function in which alterations may affect normal responses to infectious and inflammatory stimuli.


Assuntos
Proteínas de Ligação a DNA/genética , Malária Cerebral/genética , Fatores de Transcrição/genética , Tuberculose Pulmonar/genética , Animais , Encéfalo/microbiologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD8-Positivos/microbiologia , Citocinas/genética , Feminino , Inflamação/genética , Inflamação/microbiologia , Malária Cerebral/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/patogenicidade , Plasmodium berghei/patogenicidade , Tuberculose Pulmonar/microbiologia , Virulência/genética
11.
Mamm Genome ; 29(7-8): 425-445, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30167845

RESUMO

A majority of the world population is infected with herpes simplex viruses (HSV; human herpesvirus types 1 and 2). These viruses, perhaps best known for their manifestation in the genital or oral mucosa, can also cause herpes simplex encephalitis, a severe and often fatal disease of the central nervous system. Antiviral therapies for HSV are only partially effective since the virus can establish latent infections in neurons, and severe pathological sequelae in the brain are common. A better understanding of disease pathogenesis is required to develop new strategies against herpes simplex encephalitis, including the precise viral and host genetic determinants that promote virus invasion into the central nervous system and its associated immunopathology. Here we review the current understanding of herpes simplex encephalitis from the host genome perspective, which has been illuminated by groundbreaking work on rare herpes simplex encephalitis patients together with mechanistic insight from single-gene mouse models of disease. A complex picture has emerged, whereby innate type I interferon-mediated antiviral signaling is a central pathway to control viral replication, and the regulation of immunopathology and the balance between apoptosis and autophagy are critical to disease severity in the central nervous system. The lessons learned from mouse studies inform us on fundamental defense mechanisms at the interface of host-pathogen interactions within the central nervous system, as well as possible rationales for intervention against infections from severe neuropathogenic viruses.


Assuntos
Suscetibilidade a Doenças , Encefalite por Herpes Simples/etiologia , Interações Hospedeiro-Patógeno , Simplexvirus/fisiologia , Animais , Autofagia , Biomarcadores , Morte Celular , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Predisposição Genética para Doença , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Celular , Imunidade Inata , Interferon Tipo I/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Transdução de Sinais
12.
J Immunol ; 197(6): 2325-37, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27511735

RESUMO

NK cells play a major role in immune defense against human and murine CMV (MCMV) infection. Although the MCMV genome encodes for MHC class I-homologous decoy ligands for inhibitory NK cell receptors to evade detection, some mouse strains have evolved activating receptors, such as Ly49H, to recognize these ligands and initiate an immune response. In this study, we demonstrate that approximately half of the Ly49H-expressing (Ly49H(+)) NK cells in the spleen and liver of C57BL/6 mice also express the inhibitory NKR-P1B receptor. During MCMV infection, the NKR-P1B(-)Ly49H(+) NK cell subset proliferates to constitute the bulk of the NK cell population. This NK cell subset also confers better protection against MCMV infection compared with the NKR-P1B(+)Ly49H(+) subset. The two populations are composed of cells that differ in their surface expression of receptors such as Ly49C/I and NKG2A/C/E, as well as developmental markers, CD27 and CD11b, and the high-affinity IL-2R (CD25) following infection. Although the NKR-P1B(+) NK cells can produce effector molecules such as IFNs and granzymes, their proliferation is inhibited during infection. A similar phenotype in MCMV-infected Clr-b-deficient mice, which lack the ligand for NKR-P1B, suggests the involvement of ligands other than the host Clr-b. Most interestingly, genetic deficiency of the NKR-P1B, but not Clr-b, results in accelerated virus clearance and recovery from MCMV infection. This study is particularly significant because the mouse NKR-P1B:Clr-b receptor:ligand system represents the closest homolog of the human NKR-P1A:LLT1 system and may have a direct relevance to human CMV infection.


Assuntos
Infecções por Herpesviridae/imunologia , Células Matadoras Naturais/imunologia , Subfamília B de Receptores Semelhantes a Lectina de Células NK/imunologia , Animais , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Muromegalovirus/imunologia , Muromegalovirus/fisiologia , Subfamília B de Receptores Semelhantes a Lectina de Células NK/deficiência , Subfamília B de Receptores Semelhantes a Lectina de Células NK/genética
13.
J Immunol ; 193(12): 6061-9, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25392524

RESUMO

The outcome of mouse CMV (MCMV) infection varies among different inbred mouse strains depending on NK cell effector functions governed through recognition receptor triggering. NK cells from different mouse strains possess diverse repertoires of activating or inhibitory Ly49 receptors, which share some of their polymorphic MHC class I (MHC-I) ligands. By examining the NK cell response to MCMV infection in novel BALB substrains congenic for different MHC (or H-2 in mice) haplotypes, we show that recognition of viral MHC-I-like protein m157 by inhibitory Ly49C receptor allows escape from NK cell control of viral replication. Dominant inhibition by Ly49C bound to self-H-2(b) encoded MHC-I molecules masks this effect, which only becomes apparent in distinct H-2 haplotypes, such as H-2(f). The recognition of m157-expressing cells by Ly49C resulted in both decreased NK cell killing in vitro and reduced rejection in vivo. Further, control of infection with m157-deletant (Δm157) MCMV was improved in mice carrying H-2 molecules unrecognized by Ly49C but allowing expansion of NK cell effectors expressing activating Ly49L receptors. Hence, our study is the first, to our knowledge, to demonstrate that MHC-I mimicry strategies used by MCMV to avoid NK cell control are biologically relevant during in vivo viral infection. Of value for human studies is that only a few genetic assortments conditional on the repertoires of viral MHC-I-like proteins/host NK receptors/MHC haplotypes should allow efficient protection against CMV infection.


Assuntos
Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Muromegalovirus/genética , Proteínas Virais/genética , Animais , Linhagem Celular , Citotoxicidade Imunológica , Suscetibilidade a Doenças/imunologia , Feminino , Predisposição Genética para Doença , Antígenos H-2/genética , Antígenos H-2/imunologia , Infecções por Herpesviridae/genética , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Muromegalovirus/imunologia , Muromegalovirus/patogenicidade , Subfamília A de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas Virais/imunologia
14.
J Immunol ; 192(1): 259-70, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24285835

RESUMO

Salmonella enterica is a ubiquitous Gram-negative intracellular bacterium that continues to pose a global challenge to human health. The etiology of Salmonella pathogenesis is complex and controlled by pathogen, environmental, and host genetic factors. In fact, patients immunodeficient in genes in the IL-12, IL-23/IFN-γ pathway are predisposed to invasive nontyphoidal Salmonella infection. Using a forward genomics approach by N-ethyl-N-nitrosourea (ENU) germline mutagenesis in mice, we identified the Ity14 (Immunity to Typhimurium locus 14) pedigree exhibiting increased susceptibility following in vivo Salmonella challenge. A DNA-binding domain mutation (p.G418_E445) in Stat4 (Signal Transducer and Activator of Transcription Factor 4) was the causative mutation. STAT4 signals downstream of IL-12 to mediate transcriptional regulation of inflammatory immune responses. In mutant Ity14 mice, the increased splenic and hepatic bacterial load resulted from an intrinsic defect in innate cell function, IFN-γ-mediated immunity, and disorganized granuloma formation. We further show that NK and NKT cells play an important role in mediating control of Salmonella in Stat4(Ity14/Ity14) mice. Stat4(Ity14/Ity14) mice had increased expression of genes involved in cell-cell interactions and communication, as well as increased CD11b expression on a subset of splenic myeloid dendritic cells, resulting in compromised recruitment of inflammatory cells to the spleen during Salmonella infection. Stat4(Ity14/Ity14) presented upregulated compensatory mechanisms, although inefficient and ultimately Stat4(Ity14/Ity14) mice develop fatal bacteremia. The following study further elucidates the pathophysiological impact of STAT4 during Salmonella infection.


Assuntos
Regulação da Expressão Gênica , Predisposição Genética para Doença , Interferon gama/imunologia , Mutação , Fator de Transcrição STAT4/genética , Salmonelose Animal/genética , Salmonelose Animal/imunologia , Transcrição Gênica , Animais , Carga Bacteriana , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Proteínas de Transporte de Cátions/genética , Análise por Conglomerados , Análise Mutacional de DNA , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Loci Gênicos , Imunidade Inata/genética , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Fígado/imunologia , Fígado/metabolismo , Fígado/microbiologia , Camundongos , Mutação/efeitos dos fármacos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Compostos de Nitrosoureia/toxicidade , Linhagem , Salmonelose Animal/microbiologia , Salmonelose Animal/mortalidade , Salmonella typhimurium/imunologia , Baço/imunologia , Baço/metabolismo , Baço/microbiologia , Transcriptoma
15.
Infect Immun ; 83(2): 759-68, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25452553

RESUMO

We identify an N-ethyl-N-nitrosourea (ENU)-induced I23N mutation in the THEMIS protein that causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA. Themis(I23N) homozygous mice show reduced CD4(+) and CD8(+) T lymphocyte numbers. ECM resistance in P. berghei ANKA-infected Themis(I23N) mice is associated with decreased cerebral cellular infiltration, retention of blood-brain barrier integrity, and reduced proinflammatory cytokine production. THEMIS(I23N) protein expression is absent from mutant mice, concurrent with the decreased THEMIS(I23N) stability observed in vitro. Biochemical studies in vitro and functional complementation in vivo in Themis(I23N/+):Lck(-/+) doubly heterozygous mice demonstrate that functional coupling of THEMIS to LCK tyrosine kinase is required for ECM pathogenesis. Damping of proinflammatory responses in Themis(I23N) mice causes susceptibility to pulmonary tuberculosis. Thus, THEMIS is required for the development and ultimately the function of proinflammatory T cells. Themis(I23N) mice can be used to study the newly discovered association of THEMIS (6p22.33) with inflammatory bowel disease and multiple sclerosis.


Assuntos
Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Malária Cerebral/imunologia , Plasmodium berghei/imunologia , Proteínas/genética , Tuberculose Pulmonar/imunologia , Animais , Barreira Hematoencefálica , Encéfalo/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Doença Celíaca/genética , Etilnitrosoureia , Expressão Gênica , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intercelular , Malária Cerebral/parasitologia , Malária Cerebral/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parasitemia/patologia , Proteínas/imunologia , Tuberculose Pulmonar/microbiologia
16.
PLoS Pathog ; 9(4): e1003256, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23592984

RESUMO

Influenza A virus (IAV) triggers a contagious and potentially lethal respiratory disease. A protective IL-1ß response is mediated by innate receptors in macrophages and lung epithelial cells. NLRP3 is crucial in macrophages; however, which sensors elicit IL-1ß secretion in lung epithelial cells remains undetermined. Here, we describe for the first time the relative roles of the host innate receptors RIG-I (DDX58), TLR3, and NLRP3 in the IL-1ß response to IAV in primary lung epithelial cells. To activate IL-1ß secretion, these cells employ partially redundant recognition mechanisms that differ from those described in macrophages. RIG-I had the strongest effect through a MAVS/TRIM25/Riplet-dependent type I IFN signaling pathway upstream of TLR3 and NLRP3. Notably, RIG-I also activated the inflammasome through interaction with caspase 1 and ASC in primary lung epithelial cells. Thus, NS1, an influenza virulence factor that inhibits the RIG-I/type I IFN pathway, strongly modulated the IL-1ß response in lung epithelial cells and in ferrets. The NS1 protein derived from a highly pathogenic strain resulted in increased interaction with RIG-I and inhibited type I IFN and IL-1ß responses compared to the least pathogenic virus strains. These findings demonstrate that in IAV-infected lung epithelial cells RIG-I activates the inflammasome both directly and through a type I IFN positive feedback loop.


Assuntos
Proteínas de Transporte/metabolismo , RNA Helicases DEAD-box/metabolismo , Inflamassomos/metabolismo , Vírus da Influenza A Subtipo H1N1 , Interferon beta/metabolismo , Receptor 3 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/genética , Caspase 1/genética , Caspase 1/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células Epiteliais/metabolismo , Furões , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H1N1/metabolismo , Pulmão/metabolismo , Pulmão/virologia , Macrófagos/imunologia , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR , Interferência de RNA , Receptores Imunológicos , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/genética , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/metabolismo , Proteínas não Estruturais Virais/metabolismo
17.
PLoS Pathog ; 9(9): e1003637, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24068938

RESUMO

Herpes simplex encephalitis (HSE) is a lethal neurological disease resulting from infection with Herpes Simplex Virus 1 (HSV-1). Loss-of-function mutations in the UNC93B1, TLR3, TRIF, TRAF3, and TBK1 genes have been associated with a human genetic predisposition to HSE, demonstrating the UNC93B-TLR3-type I IFN pathway as critical in protective immunity to HSV-1. However, the TLR3, UNC93B1, and TRIF mutations exhibit incomplete penetrance and represent only a minority of HSE cases, perhaps reflecting the effects of additional host genetic factors. In order to identify new host genes, proteins and signaling pathways involved in HSV-1 and HSE susceptibility, we have implemented the first genome-wide mutagenesis screen in an in vivo HSV-1 infectious model. One pedigree (named P43) segregated a susceptible trait with a fully penetrant phenotype. Genetic mapping and whole exome sequencing led to the identification of the causative nonsense mutation L3X in the Receptor-type tyrosine-protein phosphatase C gene (Ptprc(L3X)), which encodes for the tyrosine phosphatase CD45. Expression of MCP1, IL-6, MMP3, MMP8, and the ICP4 viral gene were significantly increased in the brain stems of infected Ptprc(L3X) mice accounting for hyper-inflammation and pathological damages caused by viral replication. Ptprc(L3X) mutation drastically affects the early stages of thymocytes development but also the final stage of B cell maturation. Transfer of total splenocytes from heterozygous littermates into Ptprc(L3X) mice resulted in a complete HSV-1 protective effect. Furthermore, T cells were the only cell population to fully restore resistance to HSV-1 in the mutants, an effect that required both the CD4⁺ and CD8⁺ T cells and could be attributed to function of CD4⁺ T helper 1 (Th1) cells in CD8⁺ T cell recruitment to the site of infection. Altogether, these results revealed the CD45-mediated T cell function as potentially critical for infection and viral spread to the brain, and also for subsequent HSE development.


Assuntos
Códon sem Sentido , Encefalite por Herpes Simples/genética , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Imunidade Celular , Antígenos Comuns de Leucócito/metabolismo , Células Th1/imunologia , Animais , Tronco Encefálico/imunologia , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Tronco Encefálico/virologia , Células Cultivadas , Cruzamentos Genéticos , Suscetibilidade a Doenças , Encefalite por Herpes Simples/etiologia , Feminino , Estudo de Associação Genômica Ampla , Herpes Simples/patologia , Herpes Simples/fisiopatologia , Herpes Simples/virologia , Antígenos Comuns de Leucócito/genética , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutagênese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Análise de Sobrevida , Células Th1/metabolismo , Células Th1/patologia , Células Th1/virologia
18.
Hum Mutat ; 35(5): 594-600, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24616178

RESUMO

The polymorphism 677C>T (NM_005957.4:c.665C>T/p.Ala222Val, rs1801133:C>T) in methylenetetrahydrofolate reductase (MTHFR) results in mild enzymatic deficiency and increased risk for several complex traits including adverse reproductive outcomes, birth defects, and heart disease. Despite these deleterious effects, homozygosity is high (5%-15%) in many populations, and among the highest in Mediterranean regions, where malaria was historically endemic and may have conferred a selective advantage for other mutations. We infected Mthfr-deficient (Mthfr(+) (/-) ) and MTHFR overexpressing (MTHFR(Tg) ) mice with Plasmodium berghei ANKA to induce cerebral malaria. Mthfr(+/-) mice survived longer (P < 0.02, log-rank test), and MTHFR(Tg) mice died earlier (P < 0.05, log-rank test) after infection compared with wild-type littermates. Flow cytometry revealed increased lymphocyte populations and increased CCR4(+) NK cells in spleen of Mthfr(+) (/-) mice; MTHFR(Tg) animals had decreased numbers of these NK cells. Interferon-γ and interleukin-10 immunoreactive proteins were increased and decreased, respectively, in brain of Mthfr(+/-) mice compared with wild-type. We suggest that mild MTHFR deficiency protects against malarial infection and that this phenomenon may have led to the high frequency of the 677C>T/c.665C>T variant in human populations.


Assuntos
Homocistinúria/genética , Malária Cerebral/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/deficiência , Espasticidade Muscular/genética , Seleção Genética/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Ácido Fólico/genética , Homocistinúria/metabolismo , Homocistinúria/patologia , Humanos , Interferon gama/biossíntese , Interleucina-10/biossíntese , Malária Cerebral/etiologia , Malária Cerebral/patologia , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Camundongos , Espasticidade Muscular/metabolismo , Espasticidade Muscular/patologia , Plasmodium/metabolismo , Plasmodium/patogenicidade , Polimorfismo de Nucleotídeo Único , Transtornos Psicóticos/genética , Transtornos Psicóticos/metabolismo , Transtornos Psicóticos/patologia
19.
J Immunol ; 188(8): 3949-60, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22427645

RESUMO

Seasonal influenza outbreaks and recurrent influenza pandemics present major challenges to public health. By studying immunological responses to influenza in different host species, it may be possible to discover common mechanisms of susceptibility in response to various influenza strains. This could lead to novel therapeutic targets with wide clinical application. Using a mouse-adapted strain of influenza (A/HK/1/68-MA20 [H3N2]), we produced a mouse model of severe influenza that reproduces the hallmark high viral load and overexpression of cytokines associated with susceptibility to severe influenza in humans. We mapped genetic determinants of the host response using a panel of 29 closely related mouse strains (AcB/BcA panel of recombinant congenic strains) created from influenza-susceptible A/J and influenza-resistant C57BL/6J (B6) mice. Combined clinical quantitative trait loci (QTL) and lung expression QTL mapping identified candidate genes for two sex-specific QTL on chromosomes 2 and 17. The former includes the previously described Hc gene, a deficit of which is associated with the susceptibility phenotype in females. The latter includes the phospholipase gene Pla2g7 and Tnfrsf21, a member of the TNFR superfamily. Confirmation of the gene underlying the chromosome 17 QTL may reveal new strategies for influenza treatment.


Assuntos
Cromossomos de Mamíferos/genética , Vírus da Influenza A Subtipo H3N2 , Influenza Humana/genética , Locos de Características Quantitativas , 1-Alquil-2-acetilglicerofosfocolina Esterase , Alelos , Animais , Mapeamento Cromossômico , Cromossomos de Mamíferos/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Genótipo , Especificidade de Hospedeiro , Humanos , Influenza Humana/imunologia , Influenza Humana/virologia , Pulmão/imunologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Fenótipo , Fosfolipases A2/genética , Fosfolipases A2/imunologia , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Fatores Sexuais
20.
PLoS Genet ; 7(4): e1001368, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21533075

RESUMO

The cytomegalovirus resistance locus Cmv3 has been linked to an epistatic interaction between two loci: a Natural Killer (NK) cell receptor gene and the major histocompatibility complex class I (MHC-I) locus. To demonstrate the interaction between Cmv3 and H2(k), we generated double congenic mice between MA/My and BALB.K mice and an F(2) cross between FVB/N (H-2(q)) and BALB.K (H2(k)) mice, two strains susceptible to mouse cytomegalovirus (MCMV). Only mice expressing H2(k) in conjunction with Cmv3(MA/My) or Cmv3(FVB) were resistant to MCMV infection. Subsequently, an F(3) cross was carried out between transgenic FVB/H2-D(k) and MHC-I deficient mice in which only the progeny expressing Cmv3(FVB) and a single H2-D(k) class-I molecule completely controlled MCMV viral loads. This phenotype was shown to be NK cell-dependent and associated with subsequent NK cell proliferation. Finally, we demonstrated that a number of H2(q) alleles influence the expression level of H2(q) molecules, but not intrinsic functional properties of NK cells; viral loads, however, were quantitatively proportional to the number of H2(q) alleles. Our results support a model in which H-2(q) molecules convey Ly49-dependent inhibitory signals that interfere with the action of H2-D(k) on NK cell activation against MCMV infection. Thus, the integration of activating and inhibitory signals emanating from various MHC-I/NK cell receptor interactions regulates NK cell-mediated control of viral load.


Assuntos
Genes MHC Classe I , Infecções por Herpesviridae/imunologia , Muromegalovirus/imunologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/imunologia , Receptores de Células Matadoras Naturais/genética , Animais , Células Cultivadas , Frequência do Gene , Infecções por Herpesviridae/virologia , Imunidade Inata , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos , Camundongos Transgênicos , Muromegalovirus/patogenicidade , Fenótipo , Receptores de Células Matadoras Naturais/imunologia , Transgenes , Carga Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa