Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
PLoS Genet ; 12(7): e1006185, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27438086

RESUMEN

The guanine nucleotide exchange factor Vav1 is essential for transducing T cell antigen receptor signals and therefore plays an important role in T cell development and activation. Our previous genetic studies identified a locus on rat chromosome 9 that controls the susceptibility to neuroinflammation and contains a non-synonymous polymorphism in the major candidate gene Vav1. To formally demonstrate the causal implication of this polymorphism, we generated a knock-in mouse bearing this polymorphism (Vav1R63W). Using this model, we show that Vav1R63W mice display reduced susceptibility to experimental autoimmune encephalomyelitis (EAE) induced by MOG35-55 peptide immunization. This is associated with a lower production of effector cytokines (IFN-γ, IL-17 and GM-CSF) by autoreactive CD4 T cells. Despite increased proportion of Foxp3+ regulatory T cells in Vav1R63W mice, we show that this lowered cytokine production is intrinsic to effector CD4 T cells and that Treg depletion has no impact on EAE development. Finally, we provide a mechanism for the above phenotype by showing that the Vav1R63W variant has normal enzymatic activity but reduced adaptor functions. Together, these data highlight the importance of Vav1 adaptor functions in the production of inflammatory cytokines by effector T cells and in the susceptibility to neuroinflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Variación Genética , Proteínas Proto-Oncogénicas c-vav/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T Reguladores/citología , Animales , Calcio/metabolismo , Sistema Nervioso Central/fisiopatología , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Factores de Transcripción Forkhead/metabolismo , Predisposición Genética a la Enfermedad , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Polimorfismo Genético , Ratas , Transducción de Señal , Timo/metabolismo
2.
J Immunol ; 195(4): 1608-16, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26163585

RESUMEN

The development of inflammatory diseases depends on complex interactions between several genes and various environmental factors. Discovering new genetic risk factors and understanding the mechanisms whereby they influence disease development is of paramount importance. We previously reported that deficiency in Themis1, a new actor of TCR signaling, impairs regulatory T cell (Treg) function and predisposes Brown-Norway (BN) rats to spontaneous inflammatory bowel disease (IBD). In this study, we reveal that the epistasis between Themis1 and Vav1 controls the occurrence of these phenotypes. Indeed, by contrast with BN rats, Themis1 deficiency in Lewis rats neither impairs Treg suppressive functions nor induces pathological manifestations. By using congenic lines on the BN genomic background, we show that the impact of Themis1 deficiency on Treg suppressive functions depends on a 117-kb interval coding for a R63W polymorphism that impacts Vav1 expression and functions. Indeed, the introduction of a 117-kb interval containing the Lewis Vav1-R63 variant restores Treg function and protects Themis1-deficient BN rats from spontaneous IBD development. We further show that Themis1 binds more efficiently to the BN Vav1-W63 variant and is required to stabilize its recruitment to the transmembrane adaptor LAT and to fully promote the activation of Erk kinases. Together, these results highlight the importance of the signaling pathway involving epistasis between Themis1 and Vav1 in the control of Treg suppressive function and susceptibility to IBD development.


Asunto(s)
Epistasis Genética , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Proto-Oncogénicas c-vav/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Mutación , Proteínas Proto-Oncogénicas c-vav/metabolismo , Ratas , Ratas Transgénicas , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timocitos/inmunología , Timocitos/metabolismo
3.
Infect Immun ; 82(7): 2670-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24686056

RESUMEN

ALOX12 is a gene encoding arachidonate 12-lipoxygenase (12-LOX), a member of a nonheme lipoxygenase family of dioxygenases. ALOX12 catalyzes the addition of oxygen to arachidonic acid, producing 12-hydroperoxyeicosatetraenoic acid (12-HPETE), which can be reduced to the eicosanoid 12-HETE (12-hydroxyeicosatetraenoic acid). 12-HETE acts in diverse cellular processes, including catecholamine synthesis, vasoconstriction, neuronal function, and inflammation. Consistent with effects on these fundamental mechanisms, allelic variants of ALOX12 are associated with diseases including schizophrenia, atherosclerosis, and cancers, but the mechanisms have not been defined. Toxoplasma gondii is an apicomplexan parasite that causes morbidity and mortality and stimulates an innate and adaptive immune inflammatory reaction. Recently, it has been shown that a gene region known as Toxo1 is critical for susceptibility or resistance to T. gondii infection in rats. An orthologous gene region with ALOX12 centromeric is also present in humans. Here we report that the human ALOX12 gene has susceptibility alleles for human congenital toxoplasmosis (rs6502997 [P, <0.000309], rs312462 [P, <0.028499], rs6502998 [P, <0.029794], and rs434473 [P, <0.038516]). A human monocytic cell line was genetically engineered using lentivirus RNA interference to knock down ALOX12. In ALOX12 knockdown cells, ALOX12 RNA expression decreased and levels of the ALOX12 substrate, arachidonic acid, increased. ALOX12 knockdown attenuated the progression of T. gondii infection and resulted in greater parasite burdens but decreased consequent late cell death of the human monocytic cell line. These findings suggest that ALOX12 influences host responses to T. gondii infection in human cells. ALOX12 has been shown in other studies to be important in numerous diseases. Here we demonstrate the critical role ALOX12 plays in T. gondii infection in humans.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Toxoplasmosis Congénita/genética , Alelos , Araquidonato 12-Lipooxigenasa/química , Araquidonato 12-Lipooxigenasa/genética , Ácido Araquidónico/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular , Estudios de Cohortes , Citocinas/genética , Citocinas/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Variación Genética , Humanos , Masculino , Monocitos/metabolismo , Monocitos/parasitología , Plásmidos/genética , Interferencia de ARN , ARN Interferente Pequeño , Toxoplasmosis Congénita/inmunología , Toxoplasmosis Congénita/parasitología
4.
PLoS One ; 8(5): e63585, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23704921

RESUMEN

Epithelial repair following acute kidney injury (AKI) requires epithelial-mesenchyme-epithelial cycling associated with transient re-expression of genes normally expressed during kidney development as well as activation of growth factors and cytokine-induced signaling. In normal kidney, the Hnf-1ß transcription factor drives nephrogenesis, tubulogenesis and epithelial homeostasis through the regulation of epithelial planar cell polarity and expression of developmental or tubular segment-specific genes. In a mouse model of ischemic AKI induced by a 2-hours hemorrhagic shock, we show that expression of this factor is tightly regulated in the early phase of renal repair with a biphasic expression profile (early down-regulation followed by transient over-expression). These changes are associated to tubular epithelial differentiation as assessed by KSP-cadherin and megalin-cubilin endocytic complex expression analysis. In addition, early decrease in Hnf1b expression is associated with the transient over-expression of one of its main target genes, the suppressor of cytokine signaling Socs3, which has been shown essential for renal repair. In vitro, hypoxia induced early up-regulation of Hnf-1ß from 1 to 24 hours, independently of the hypoxia-inducible factor Hif-1α. When prolonged, hypoxia induced Hnf-1ß down-regulation while normoxia led to Hnf-1ß normalization. Last, Hnf-1ß down-regulation using RNA interference in HK-2 cells led to phenotype switch from an epithelial to a mesenchyme state. Taken together, we showed that Hnf-1ß may drive recovery from ischemic AKI by regulating both the expression of genes important for homeostasis control during organ repair and the state of epithelial cell differentiation.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/patología , Factor Nuclear 1-beta del Hepatocito/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/metabolismo , Riñón/patología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Biomarcadores/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Línea Celular , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Factor Nuclear 1-beta del Hepatocito/antagonistas & inhibidores , Factor Nuclear 1-beta del Hepatocito/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Riñón/efectos de los fármacos , Riñón/fisiopatología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Ratones , Ratones Endogámicos C57BL , Inhibidores de Prolil-Hidroxilasa/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Choque Hemorrágico/complicaciones , Choque Hemorrágico/patología , Choque Hemorrágico/fisiopatología , Cicatrización de Heridas/efectos de los fármacos
5.
Nephron Clin Pract ; 120(2): c71-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22269832

RESUMEN

BACKGROUND/AIMS: HNF1B nephropathy is characterized by dominantly inherited renal hypodysplasia with few cysts, slow renal decline and hypomagnesemia. Mice with antenatal inactivation of HNF1B are characterized by polycystic kidneys, renal failure and a profound decrease in cystic gene (Pkhd1, Umod, Pkd2) expression. Mice with inactivation after postnatal day 10 have no renal phenotype. METHODS: Quantification of mRNA expression of HNF1B, six of its potential target genes (PKHD1, PKD1, PKD2, IFT88, TMEM27 and UMOD) and three genes involved in the Mg(2+) renal homeostasis (ATP1A1, FXYD2 and CLDN16) in the urinary sediment of 11 individuals with mutation of HNF1B and in 9 controls (non-invasive assessment of the renal transcriptome). RESULTS: As compared to controls, no difference was observed in the urinary mRNA amount of HNF1B and the renal cystic genes. A significant increase in the expression of ATP1A1, which encodes the α1-subunit of the Na(+)/K(+)-ATPase, was identified in HNF1B patients consistent with its role in Mg(2+) homeostasis. CONCLUSION: Assessment of mRNA expression in urinary sediment is a non-invasive method applicable to gain insights into the pathophysiology of inherited nephropathies in humans. HNF1B nephropathy is generally not associated with postnatal down-expression of renal cystic genes in human, a finding consistent with mouse models.


Asunto(s)
Expresión Génica/genética , Factor Nuclear 1-beta del Hepatocito/genética , Enfermedades Renales Quísticas/genética , Mutación/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Francia , Humanos , Enfermedades Renales Quísticas/orina , Masculino , Persona de Mediana Edad , ATPasa Intercambiadora de Sodio-Potasio/genética , Adulto Joven
6.
J Exp Med ; 208(11): 2183-91, 2011 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-21948080

RESUMEN

CD4(+) regulatory T cells (T(reg) cells) expressing the transcription factor Foxp3 play a pivotal role in maintaining peripheral tolerance by inhibiting the expansion and function of pathogenic conventional T cells (T(conv) cells). In this study, we show that a locus on rat chromosome 9 controls the size of the natural T(reg) cell compartment. Fine mapping of this locus with interval-specific congenic lines and association experiments using single nucleotide polymorphisms (SNPs) identified a nonsynonymous SNP in the Vav1 gene that leads to the substitution of an arginine by a tryptophan (p.Arg63Trp). This p.Arg63Trp polymorphism is associated with increased proportion and absolute numbers of T(reg) cells in the thymus and peripheral lymphoid organs, without impacting the size of the T(conv) cell compartment. This polymorphism is also responsible for Vav1 constitutive activation, revealed by its tyrosine 174 hyperphosphorylation and increased guanine nucleotide exchange factor activity. Moreover, it induces a marked reduction in Vav1 cellular contents and a reduction of Ca(2+) flux after TCR engagement. Together, our data reveal a key role for Vav1-dependent T cell antigen receptor signaling in natural T(reg) cell development.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Polimorfismo Genético , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Linfocitos T Reguladores/fisiología , Animales , Animales Congénicos , Arginina/genética , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/fisiología , Células Cultivadas , Cromosomas de los Mamíferos/genética , Factores de Transcripción Forkhead/genética , Células HEK293 , Humanos , Ratas , Ratas Endogámicas Lew , Linfocitos T Reguladores/citología , Quimera por Trasplante , Triptófano/genética
7.
Sci Transl Med ; 1(10): 10ra21, 2009 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20368159

RESUMEN

Multiple sclerosis, the most common cause of progressive neurological disability in young adults, is a chronic inflammatory disease. There is solid evidence for a genetic influence in multiple sclerosis, and deciphering the causative genes could reveal key pathways influencing the disease. A genome region on rat chromosome 9 regulates experimental autoimmune encephalomyelitis, a model for multiple sclerosis. Using interval-specific congenic rat lines and association of single-nucleotide polymorphisms with inflammatory phenotypes, we localized the gene of influence to Vav1, which codes for a signal-transducing protein in leukocytes. Analysis of seven human cohorts (12,735 individuals) demonstrated an association of rs2546133-rs2617822 haplotypes in the first VAV1 intron with multiple sclerosis (CA: odds ratio, 1.18; CG: odds ratio, 0.86; TG: odds ratio, 0.90). The risk CA haplotype also predisposed for higher VAV1 messenger RNA expression. VAV1 expression was increased in individuals with multiple sclerosis and correlated with tumor necrosis factor and interferon-gamma expression in peripheral blood and cerebrospinal fluid cells. We conclude that VAV1 plays a central role in controlling central nervous system immune-mediated disease and proinflammatory cytokine production critical for disease pathogenesis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/fisiopatología , Esclerosis Múltiple/fisiopatología , Proteínas Proto-Oncogénicas c-vav/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Interferón gamma/genética , Esclerosis Múltiple/inmunología , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-vav/genética , Sitios de Carácter Cuantitativo , Ratas , Factor de Necrosis Tumoral alfa/genética
8.
J Immunol ; 177(4): 2285-93, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16887989

RESUMEN

Lat(Y136F) knock-in mice harbor a point mutation in Tyr(136) of the linker for activation of T cells and show accumulation of Th2 effector cells and IgG1 and IgE hypergammaglobulinemia. B cell activation is not a direct effect of the mutation on B cells since in the absence of T cells, mutant B cells do not show an activated phenotype. After adoptive transfer of linker for activation of T cell mutant T cells into wild-type, T cell-deficient recipients, recipient B cells become activated. We show in vivo and in vitro that the Lat(Y136F) mutation promotes T cell-dependent B cell activation leading to germinal center, memory, and plasma cell formation even in an MHC class II-independent manner. All the plasma and memory B cell populations found in physiological T cell-dependent B cell responses are found. Characterization of the abundant plasmablasts found in secondary lymphoid organs of Lat(Y136F) mice revealed the presence of a previously uncharacterized CD93-expressing subpopulation, whose presence was confirmed in wild-type mice after immunization. In Lat(Y136F) mice, B cell activation was polyclonal and not Ag-driven because the increase in serum IgG1 and IgE concentrations involved Abs and autoantibodies with different specificities equally. Although the noncomplement-fixing IgG1 and IgE are the only isotypes significantly increased in Lat(Y136F) serum, we observed early-onset systemic autoimmunity with nephritis showing IgE autoantibody deposits and severe proteinuria. These results show that Th2 cells developing in Lat(Y136F) mice can trigger polyclonal B cell activation and thereby lead to systemic autoimmune disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Sustitución de Aminoácidos , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Activación de Linfocitos/genética , Trastornos Linfoproliferativos/genética , Proteínas de la Membrana/fisiología , Fosfoproteínas/fisiología , Células Th2/patología , Proteínas Adaptadoras Transductoras de Señales/genética , Sustitución de Aminoácidos/genética , Animales , Enfermedades Autoinmunes/patología , Linfocitos B/citología , Linfocitos B/patología , Células Clonales , Trastornos Linfoproliferativos/inmunología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Fenilalanina/genética , Fosfoproteínas/genética , Células Th2/inmunología , Tirosina/genética
9.
Infect Immun ; 73(10): 6990-7, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16177379

RESUMEN

Toxoplasmosis is a ubiquitous parasitic infection causing a wide spectrum of diseases. It is usually asymptomatic but can lead to severe ocular and neurological disorders. Among the small-animal models available to study factors that determine susceptibility to toxoplasmosis, the rat appears to be rather similar to humans, particularly in terms of resistance to acute infection. Here, we demonstrate that the Lewis (LEW) rat strain displays an unexpected refractoriness to Toxoplasma infection. Complete resistance was assessed by both negative anti-Toxoplasma serology and lack of detection of the parasite during the course of infection. In this model, sex, age, major histocompatibility complex, and inoculum size had no effect on resistance. Interestingly, progeny from F(1) hybrid crosses between Fischer (F344) or Brown Norway susceptible rats and LEW resistant rats were also fully resistant, showing a dominant effect of the gene or set of genes. Furthermore, resistance of the LEW rat was shown to be dependent on hematopoietic cells and partially abrogated by neutralization of endogenous gamma interferon. To our knowledge, this is the first observation of a rodent strain that is refractory to Toxoplasma infection. This model is therefore an attractive and powerful tool to dissect host genetic factors involved in susceptibility to toxoplasmosis.


Asunto(s)
Predisposición Genética a la Enfermedad , Células Madre Hematopoyéticas/inmunología , Inmunidad Innata/genética , Ratas Endogámicas Lew/genética , Toxoplasmosis Animal/genética , Toxoplasmosis Animal/inmunología , Animales , Encéfalo/parasitología , Femenino , Haplotipos , Células Madre Hematopoyéticas/metabolismo , Antígenos de Histocompatibilidad/genética , Inmunoglobulina G/sangre , Interferón gamma/metabolismo , Intestinos/parasitología , Masculino , Ratas , Ratas Endogámicas Lew/inmunología
10.
J Immunol ; 173(5): 3140-7, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15322174

RESUMEN

Differential cytokine production by T cells plays an important role in the outcome of the immune response. We show that the level of CD45RC expression differentiates rat CD8 T cells in two subpopulations, CD45RC(high) and CD45RC(low), that have different cytokine profiles and functions. Upon in vitro stimulation, in an Ag-presenting cell-independent system, CD45RC(high) CD8 T cells produce IL-2 and IFN-gamma while CD45RC(low) CD8 T cells produce IL-4, IL-10, and IL-13. In vitro, these subsets also exhibit different cytotoxic and suppressive functions. The CD45RC(high)/CD45RC(low) CD8 T cell ratio was determined in Lewis (LEW) and Brown-Norway (BN) rats. These two rat strains differ with respect to the Th1/Th2 polarization of their immune responses and to their susceptibility to develop distinct immune diseases. The CD45RC(high)/CD45RC(low) CD8 T cell ratio is higher in LEW than in BN rats, and this difference is dependent on hemopoietic cells. Linkage analysis in a F(2)(LEW x BN) intercross identified two quantitative trait loci on chromosomes 9 and 20 controlling the CD45RC(high)/CD45RC(low) CD8 T cell ratio. This genetic control was confirmed in congenic rats. The region on chromosome 9 was narrowed down to a 1.2-cM interval that was found to also control the IgE response in a model of Th2-mediated disorder. Identification of genes that control the CD45RC(high)/CD45RC(low) CD8 T cell subsets in these regions could be of great interest for the understanding of the pathophysiology of immune-mediated diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Antígenos Comunes de Leucocito/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Células Madre Hematopoyéticas/inmunología , Inmunoglobulina E/genética , Inmunoglobulina E/inmunología , Antígenos Comunes de Leucocito/genética , Masculino , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA