Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(3): 113800, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38386559

RESUMO

Infection of mice by mouse cytomegalovirus (MCMV) triggers activation and expansion of Ly49H+ natural killer (NK) cells, which are virus specific and considered to be "adaptive" or "memory" NK cells. Here, we find that signaling lymphocytic activation molecule family receptors (SFRs), a group of hematopoietic cell-restricted receptors, are essential for the expansion of Ly49H+ NK cells after MCMV infection. This activity is largely mediated by CD48, an SFR broadly expressed on NK cells and displaying augmented expression after MCMV infection. It is also dependent on the CD48 counter-receptor, 2B4, expressed on host macrophages. The 2B4-CD48 axis promotes expansion of Ly49H+ NK cells by repressing their phagocytosis by virus-activated macrophages through inhibition of the pro-phagocytic integrin lymphocyte function-associated antigen-1 (LFA-1) on macrophages. These data identify key roles of macrophages and the 2B4-CD48 pathway in controlling the expansion of adaptive NK cells following MCMV infection. Stimulation of the 2B4-CD48 axis may be helpful in enhancing adaptive NK cell responses for therapeutic purposes.


Assuntos
Infecções por Citomegalovirus , Receptores Imunológicos , Animais , Camundongos , Receptores Imunológicos/metabolismo , Antígeno CD48/metabolismo , Antígenos CD/metabolismo , Ativação Linfocitária , Células Matadoras Naturais , Receptores de Superfície Celular/metabolismo , Proteínas de Transporte/metabolismo , Macrófagos/metabolismo , Fagocitose
2.
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
3.
Front Immunol ; 12: 664218, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867937

RESUMO

Group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of early type 2 immune responses. Upon tissue damage, ILC2s are activated by alarmins such as IL-33 and rapidly secrete large amounts of type 2 signature cytokines. ILC2 activation is governed by a network of transcriptional regulators including nuclear factor (NF)-κB family transcription factors. While it is known that activating IL-33 receptor signaling results in downstream NF-κB activation, the underlying molecular mechanisms remain elusive. Here, we found that the NF-κB subunit c-Rel is required to mount effective innate pulmonary type 2 immune responses. IL-33-mediated activation of ILC2s in vitro as well as in vivo was found to induce c-Rel mRNA and protein expression. In addition, we demonstrate that IL-33-mediated activation of ILC2s leads to nuclear translocation of c-Rel in pulmonary ILC2s. Although c-Rel was found to be a critical mediator of innate pulmonary type 2 immune responses, ILC2-intrinsic deficiency of c-Rel did not have an impact on the developmental capacity of ILC2s nor affected homeostatic numbers of lung-resident ILC2s at steady state. Moreover, we demonstrate that ILC2-intrinsic deficiency of c-Rel alters the capacity of ILC2s to upregulate the expression of ICOSL and OX40L, key stimulatory receptors, and the expression of type 2 signature cytokines IL-5, IL-9, IL-13, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Collectively, our data using Rel-/- mice suggest that c-Rel promotes acute ILC2-driven allergic airway inflammation and suggest that c-Rel may contribute to the pathophysiology of ILC2-mediated allergic airway disease. It thereby represents a promising target for the treatment of allergic asthma, and evaluating the effect of established c-Rel inhibitors in this context would be of great clinical interest.


Assuntos
Imunidade Inata , Pulmão/imunologia , Subpopulações de Linfócitos/imunologia , Proteínas Proto-Oncogênicas c-rel/imunologia , Animais , Asma/imunologia , Asma/patologia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Técnicas In Vitro , Interleucina-33/imunologia , Pulmão/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/deficiência , Proteínas Proto-Oncogênicas c-rel/genética
4.
Sci Rep ; 11(1): 21171, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34707143

RESUMO

Herpes simplex virus type 1 (HSV-1) is the predominant cause of herpes simplex encephalitis (HSE), a condition characterized by acute inflammation and viral replication in the brain. Host genetics contribute to HSE onset, including monogenic defects in type I interferon signaling in cases of childhood HSE. Mouse models suggest a further contribution of immune cell-mediated inflammation to HSE pathogenesis. We have previously described a truncating mutation in the c-Rel transcription factor (RelC307X) that drives lethal HSE in 60% of HSV-1-infected RelC307X mice. In this study, we combined dual host-virus RNA sequencing with flow cytometry to explore cell populations and mechanisms involved in RelC307X-driven HSE. At day 5 postinfection, prior to HSE clinical symptom onset, elevated HSV-1 transcription was detected together with augmented host interferon-stimulated and inflammatory gene expression in the brainstems of high-responding RelC307X mice, predictive of HSE development. This early induction of host gene expression preceded pathological infiltration of myeloid and T cells in RelC307X mice at HSE onset by day 7. Thus, we establish c-Rel as an early regulator of viral and host responses during mouse HSE. These data further highlight the importance of achieving a balanced immune response and avoiding excess interferon-driven inflammation to promote HSE resistance.


Assuntos
Encefalite por Herpes Simples/metabolismo , Interferon Tipo I/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Animais , Encefalite por Herpes Simples/virologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas Proto-Oncogênicas c-rel/genética , Transdução de Sinais , Simplexvirus/genética , Simplexvirus/patogenicidade , Simplexvirus/fisiologia , Linfócitos T/metabolismo , Linfócitos T/virologia
5.
Nat Microbiol ; 4(9): 1516-1531, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31285585

RESUMO

Salmonella presents a global public health concern. Central to Salmonella pathogenicity is an ability to subvert host defences through strategically targeting host proteins implicated in restricting infection. Therefore, to gain insight into the host-pathogen interactions governing Salmonella infection, we performed an in vivo genome-wide mutagenesis screen to uncover key host defence proteins. This revealed an uncharacterized role of CYRI (FAM49B) in conferring host resistance to Salmonella infection. We show that CYRI binds to the small GTPase RAC1 through a conserved domain present in CYFIP proteins, which are known RAC1 effectors that stimulate actin polymerization. However, unlike CYFIP proteins, CYRI negatively regulates RAC1 signalling, thereby attenuating processes such as macropinocytosis, phagocytosis and cell migration. This enables CYRI to counteract Salmonella at various stages of infection, including bacterial entry into non-phagocytic and phagocytic cells as well as phagocyte-mediated bacterial dissemination. Intriguingly, to dampen its effects, the bacterial effector SopE, a RAC1 activator, selectively targets CYRI following infection. Together, this outlines an intricate host-pathogen signalling interplay that is crucial for determining bacterial fate. Notably, our study also outlines a role for CYRI in restricting infection mediated by Mycobacterium tuberculosis and Listeria monocytogenes. This provides evidence implicating CYRI cellular functions in host defence beyond Salmonella infection.


Assuntos
Infecções Bacterianas/prevenção & controle , Citoesqueleto/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Mitocondriais/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Carga Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Citoesqueleto/genética , Resistência à Doença/genética , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Listeria monocytogenes/metabolismo , Listeria monocytogenes/fisiologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Proteínas Mitocondriais/genética , Mutação , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/fisiologia , Fagocitose , Ligação Proteica , Salmonella typhimurium/metabolismo , Salmonella typhimurium/fisiologia , Análise de Sobrevida
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.
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
8.
Sci Rep ; 8(1): 8446, 2018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29855523

RESUMO

Activation of the inflammasome pathway is crucial for effective intracellular host defense. The mitochondrial network plays an important role in inflammasome regulation but the mechanisms linking mitochondrial homeostasis to attenuation of inflammasome activation are not fully understood. Here, we report that the Parkinson's disease-associated mitochondrial serine protease HtrA2 restricts the activation of ASC-dependent NLRP3 and AIM2 inflammasomes, in a protease activity-dependent manner. Consistently, disruption of the protease activity of HtrA2 results in exacerbated NLRP3 and AIM2 inflammasome responses in macrophages ex vivo and systemically in vivo. Mechanistically, we show that the HtrA2 protease activity regulates autophagy and controls the magnitude and duration of inflammasome signaling by preventing prolonged accumulation of the inflammasome adaptor ASC. Our findings identify HtrA2 as a non-redundant mitochondrial quality control effector that keeps NLRP3 and AIM2 inflammasomes in check.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Serina Peptidase 2 de Requerimento de Alta Temperatura A/metabolismo , Inflamassomos/metabolismo , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Autofagia , Células da Medula Óssea/citologia , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/deficiência , Caspase 1/genética , Caspase 1/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Serina Peptidase 2 de Requerimento de Alta Temperatura A/deficiência , Serina Peptidase 2 de Requerimento de Alta Temperatura A/genética , Proteínas Inibidoras de Apoptose/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores
9.
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
10.
PLoS One ; 10(9): e0138222, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26375467

RESUMO

Coxsackievirus type B3 (CVB3) is a cardiotropic enterovirus. Infection causes cardiomyocyte necrosis and myocardial inflammation. The damaged tissue that results is replaced with fibrotic or calcified tissue, which can lead to permanently altered cardiac function. The extent of pathogenesis among individuals exposed to CVB3 is dictated by a combination of host genetics, viral virulence, and the environment. Here, we aimed to identify genes that modulate cardiopathology following CVB3 infection. 129S1 mice infected with CVB3 developed increased cardiac pathology compared to 129X1 substrain mice despite no difference in viral burden. Linkage analysis identified a major locus on chromosome 7 (LOD: 8.307, P<0.0001) that controlled the severity of cardiac calcification and necrosis following infection. Sub-phenotyping and genetic complementation assays identified Abcc6 as the underlying gene. Microarray expression profiling identified genotype-dependent regulation of genes associated with mitochondria. Electron microscopy examination showed elevated deposition of hydroxyapatite-like material in the mitochondrial matrices of infected Abcc6 knockout (Abcc6-/-) mice but not in wildtype littermates. Cyclosporine A (CsA) inhibits mitochondrial permeability transition pore opening by inhibiting cyclophilin D (CypD). Treatment of Abcc6 -/- mice with CsA reduced cardiac necrosis and calcification by more than half. Furthermore, CsA had no effect on the CVB3-induced phenotype of doubly deficient CypD-/-Abcc6-/- mice. Altogether, our work demonstrates that mutations in Abcc6 render mice more susceptible to cardiac calcification following CVB3 infection. Moreover, we implicate CypD in the control of cardiac necrosis and calcification in Abcc6-deficient mice, whereby CypD inhibition is required for cardioprotection.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Calcinose/tratamento farmacológico , Infecções por Coxsackievirus/tratamento farmacológico , Ciclosporina/farmacologia , Enterovirus Humano B/efeitos dos fármacos , Inflamação/tratamento farmacológico , Miocardite/tratamento farmacológico , Animais , Calcinose/patologia , Calcinose/virologia , Infecções por Coxsackievirus/patologia , Infecções por Coxsackievirus/virologia , Peptidil-Prolil Isomerase F , Ciclofilinas/metabolismo , Feminino , Imunossupressores/farmacologia , Inflamação/patologia , Inflamação/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Proteínas de Transporte da Membrana Mitocondrial/efeitos dos fármacos , Poro de Transição de Permeabilidade Mitocondrial , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Miocardite/patologia , Miocardite/virologia , Necrose
11.
J Innate Immun ; 7(3): 315-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25675947

RESUMO

Coxsackievirus strain B serotype 3 (CVB3)-induced myocarditis is an important human disease that causes permanent tissue damage and can lead to death from acute infection or long-term morbidity caused by chronic inflammation. The timing and magnitude of immune activation following CVB3 infection can mediate a positive host outcome or increase tissue pathology. To better elucidate the role of endosomal Toll-like receptor (TLR) signaling in acute CVB3 infection, we studied mice with a loss-of-function mutation, known as Letr for 'loss of endosomal TLR response', in Unc93b1, which is a chaperone protein for TLR3, TLR7 and TLR9. Using Unc93b1(Letr/)(Letr) mice, we determined that Unc93b1-dependent TLR activation was essential for the survival of acute CVB3-induced myocarditis. We also determined that a lack of endosomal TLR signaling was associated with a higher viral load in target organs and that it increased inflammation, necrosis and fibrosis in cardiac tissue. Loss of Unc93b1 function was also associated with increased cardiac expression of Ifn-b and markers of tissue injury and fibrosis including Lcn2 and Serpina3n early after CVB3 infection. These observations establish a significant role for Unc93b1 in the regulation of the host inflammatory response to CVB3 infection and also reveal potential mediators of host tissue damage that merit further investigation in acute viral myocarditis.


Assuntos
Infecções por Coxsackievirus/imunologia , Endossomos/imunologia , Enterovirus Humano B/imunologia , Proteínas de Membrana Transportadoras/imunologia , Miocardite/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Proteínas de Fase Aguda/imunologia , Animais , Infecções por Coxsackievirus/patologia , Endossomos/patologia , Células HeLa , Humanos , Interferon beta/imunologia , Lipocalina-2 , Lipocalinas/imunologia , Camundongos , Miocardite/patologia , Proteínas Oncogênicas/imunologia , Serpinas/imunologia
12.
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
13.
J Exp Med ; 211(13): 2519-35, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25403443

RESUMO

We used a genome-wide screen in mutagenized mice to identify genes which inactivation protects against lethal neuroinflammation during experimental cerebral malaria (ECM). We identified an ECM-protective mutation in coiled-coil domain containing protein 88b (Ccdc88b), a poorly annotated gene that is found expressed specifically in spleen, bone marrow, lymph nodes, and thymus. The CCDC88B protein is abundantly expressed in immune cells, including both CD4(+) and CD8(+) T lymphocytes, and in myeloid cells, and loss of CCDC88B protein expression has pleiotropic effects on T lymphocyte functions, including impaired maturation in vivo, significantly reduced activation, reduced cell division as well as impaired cytokine production (IFN-γ and TNF) in response to T cell receptor engagement, or to nonspecific stimuli in vitro, and during the course of P. berghei infection in vivo. This identifies CCDC88B as a novel and important regulator of T cell function. The human CCDC88B gene maps to the 11q13 locus that is associated with susceptibility to several inflammatory and auto-immune disorders. Our findings strongly suggest that CCDC88B is the morbid gene underlying the pleiotropic effect of the 11q13 locus on inflammation.


Assuntos
Proteínas de Transporte/genética , Diferenciação Celular , Inflamação/imunologia , Inflamação/patologia , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Cromossomos Humanos Par 11/genética , Resistência à Doença/imunologia , Etilnitrosoureia , Feminino , Regulação da Expressão Gênica , Estudos de Associação Genética , Sistema Hematopoético/metabolismo , Humanos , Ativação Linfocitária/imunologia , Malária Cerebral/genética , Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Malária Cerebral/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dados de Sequência Molecular , Mutação/genética , Células Mieloides/metabolismo , Especificidade de Órgãos/genética , Plasmodium berghei , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo
14.
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
15.
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
16.
Cell Host Microbe ; 15(1): 23-35, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24439895

RESUMO

Cellular inhibitors of apoptosis proteins (cIAPs) are essential regulators of cell death and immunity. The corresponding contributions of IAPs to infectious disease outcomes are relatively unexplored. We find that mice deficient in cIAP2 exhibit increased susceptibility and mortality to influenza A virus infection. The lethality was not due to impaired antiviral immune functions, but rather because of death-receptor-induced programmed necrosis of airway epithelial cells that led to severe bronchiole epithelial degeneration, despite control of viral replication. Pharmacological inhibition of RIPK1 or genetic deletion of Ripk3, both kinases involved in programmed necrosis, rescued cIAP2-deficient mice from influenza-induced lethality. Genetic deletion of the death receptor agonists Fas ligand or TRAIL from the hematopoietic compartment also reversed the susceptibility of cIAP2-deficient mice. Thus, cIAP2-dependent antagonism of RIPK3-mediated programmed necrosis critically protects the host from influenza infection through maintenance of pulmonary tissue homeostasis rather than through pathogen control by the immune system.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Proteínas Inibidoras de Apoptose/imunologia , Pulmão/imunologia , Necrose/imunologia , Infecções por Orthomyxoviridae/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Mucosa Respiratória/imunologia , Animais , Proteína 3 com Repetições IAP de Baculovírus , Proteína Ligante Fas/deficiência , Proteína Ligante Fas/genética , Proteína Ligante Fas/imunologia , Regulação da Expressão Gênica , Homeostase/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Proteínas Inibidoras de Apoptose/deficiência , Proteínas Inibidoras de Apoptose/genética , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Knockout , Necrose/complicações , Necrose/genética , Necrose/mortalidade , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/mortalidade , Inibidores de Proteínas Quinases/farmacologia , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Transdução de Sinais , Análise de Sobrevida , Ligante Indutor de Apoptose Relacionado a TNF/deficiência , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Ubiquitina-Proteína Ligases
17.
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
18.
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
19.
PLoS One ; 8(2): e55331, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23390527

RESUMO

Salmonella, a ubiquitous Gram-negative intracellular bacterium, is a food borne pathogen that infects a broad range of hosts. Infection with Salmonella Typhimurium in mice is a broadly recognized experimental model resembling typhoid fever in humans. Using a N-ethyl-N-nitrosurea (ENU) mutagenesis recessive screen, we report the identification of Ity16 (Immunity to Typhimurium locus 16), a locus responsible for increased susceptibility to infection. The position of Ity16 was refined on chromosome 8 and a nonsense mutation was identified in the ankyrin 1 (Ank1) gene. ANK1 plays an important role in the formation and stabilization of the red cell cytoskeleton. The Ank1(Ity16/Ity16) mutation causes severe hemolytic anemia in uninfected mice resulting in splenomegaly, hyperbilirubinemia, jaundice, extramedullary erythropoiesis and iron overload in liver and kidneys. Ank1(Ity16/Ity16) mutant mice demonstrated low levels of hepcidin (Hamp) expression and significant increases in the expression of the growth differentiation factor 15 (Gdf15), erythropoietin (Epo) and heme oxygenase 1 (Hmox1) exacerbating extramedullary erythropoiesis, tissue iron deposition and splenomegaly. As the infection progresses in Ank1(Ity16/Ity16), the anemia worsens and bacterial load were high in liver and kidneys compared to wild type mice. Heterozygous Ank1(+/Ity16) mice were also more susceptible to Salmonella infection although to a lesser extent than Ank1(Ity16/Ity16) and they did not inherently present anemia and splenomegaly. During infection, iron accumulated in the kidneys of Ank1(+/Ity16) mice where bacterial loads were high compared to littermate controls. The critical role of HAMP in the host response to Salmonella infection was validated by showing increased susceptibility to infection in Hamp-deficient mice and significant survival benefits in Ank1(+/Ity16) heterozygous mice treated with HAMP peptide. This study illustrates that the regulation of Hamp and iron balance are crucial in the host response to Salmonella infection in Ank1 mutants.


Assuntos
Anemia Hemolítica/genética , Anquirinas/genética , Peptídeos Catiônicos Antimicrobianos/genética , Códon sem Sentido/efeitos dos fármacos , Etilnitrosoureia/toxicidade , Sobrecarga de Ferro/genética , Infecções por Salmonella/genética , Anemia Hemolítica/metabolismo , Anemia Hemolítica/microbiologia , Anemia Hemolítica/mortalidade , Animais , Anquirinas/metabolismo , Peptídeos Catiônicos Antimicrobianos/deficiência , Eritrócitos/metabolismo , Eritrócitos/patologia , Eritropoetina/genética , Eritropoetina/metabolismo , Expressão Gênica , Predisposição Genética para Doença , Fator 15 de Diferenciação de Crescimento/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Hepcidinas , Heterozigoto , Homozigoto , Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/microbiologia , Sobrecarga de Ferro/mortalidade , Fígado/metabolismo , Fígado/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Infecções por Salmonella/mortalidade , Salmonella typhimurium/fisiologia , Análise de Sobrevida
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA