Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Immunity ; 44(6): 1379-91, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27287409

RESUMEN

Two zoonotic coronaviruses (CoVs)-SARS-CoV and MERS-CoV-have crossed species to cause severe human respiratory disease. Here, we showed that induction of airway memory CD4(+) T cells specific for a conserved epitope shared by SARS-CoV and MERS-CoV is a potential strategy for developing pan-coronavirus vaccines. Airway memory CD4(+) T cells differed phenotypically and functionally from lung-derived cells and were crucial for protection against both CoVs in mice. Protection was dependent on interferon-γ and required early induction of robust innate and virus-specific CD8(+) T cell responses. The conserved epitope was also recognized in SARS-CoV- and MERS-CoV-infected human leukocyte antigen DR2 and DR3 transgenic mice, indicating potential relevance in human populations. Additionally, this epitope was cross-protective between human and bat CoVs, the progenitors for many human CoVs. Vaccine strategies that induce airway memory CD4(+) T cells targeting conserved epitopes might have broad applicability in the context of new CoVs and other respiratory virus outbreaks.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Coronavirus/inmunología , Sistema Respiratorio/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Vacunas Virales/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Células Cultivadas , Reacciones Cruzadas , Epítopos de Linfocito T/inmunología , Humanos , Inmunidad , Memoria Inmunológica , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos , Vacunación , Virión/inmunología
2.
J Immunol ; 198(11): 4413-4424, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28468970

RESUMEN

Mature peripheral double negative T (DNT) cells expressing αß TCR but lacking CD4/CD8 coreceptors play protective as well as pathogenic roles. To better understand their development and functioning in vivo, we concomitantly inactivated CD4 and CD8 genes in mice with intact MHC class I and class II molecules with the hypothesis that this would enable the development of DNT cells. We also envisaged that these DNT cells could be activated by bacterial superantigens in vivo as activation of T cells by superantigens does not require CD4 and CD8 coreceptors. Because HLA class II molecules present superantigens more efficiently than murine MHC class II molecules, CD4 CD8 double knockout (DKO) mice transgenically expressing HLA-DR3 or HLA-DQ8 molecules were generated. Although thymic cellularity was comparable between wild type (WT) and DKO mice, CD3+ αß TCR+ thymocytes were significantly reduced in DKO mice, implying defects in thymic-positive selection. Splenic CD3+ αß TCR+ cells and Foxp3+ T regulatory cells were present in DKO mice but significantly reduced. However, the in vivo inflammatory responses and immunopathology elicited by acute challenge with the staphylococcal superantigen enterotoxin B were comparable between WT and DKO mice. Choric exposure to staphylococcal enterotoxin B precipitated a lupus-like inflammatory disease with characteristic lympho-monocytic infiltration in lungs, livers, and kidneys, along with production of anti-nuclear Abs in DKO mice as in WT mice. Overall, our results suggest that DNT cells can develop efficiently in vivo and chronic exposure to bacterial superantigens may precipitate a lupus-like autoimmune disease through activation of DNT cells.


Asunto(s)
Antígenos CD4/genética , Antígenos CD4/inmunología , Antígenos CD8/genética , Antígenos CD8/inmunología , Enterotoxinas/inmunología , Superantígenos/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/inmunología , Antígeno HLA-DR3/genética , Antígeno HLA-DR3/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Bazo/citología , Bazo/inmunología , Timo/citología , Timo/inmunología
3.
J Biol Chem ; 291(8): 4079-90, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26703475

RESUMEN

We previously showed that an HLA-DR variant containing arginine at position 74 of the DRß1 chain (DRß1-Arg74) is the specific HLA class II variant conferring risk for autoimmune thyroid diseases (AITD). We also identified 5 thyroglobulin (Tg) peptides that bound to DRß1-Arg74. We hypothesized that blocking the binding of these peptides to DRß1-Arg74 could block the continuous T-cell activation in thyroiditis needed to maintain the autoimmune response to the thyroid. The aim of the current study was to identify small molecules that can block T-cell activation by Tg peptides presented within DRß1-Arg74 pockets. We screened a large and diverse library of compounds and identified one compound, cepharanthine that was able to block peptide binding to DRß1-Arg74. We then showed that Tg.2098 is the dominant peptide when inducing experimental autoimmune thyroiditis (EAT) in NOD mice expressing human DRß1-Arg74. Furthermore, cepharanthine blocked T-cell activation by thyroglobulin peptides, in particular Tg.2098 in mice that were induced with EAT. For the first time we identified a small molecule that can block Tg peptide binding and presentation to T-cells in autoimmune thyroiditis. If confirmed cepharanthine could potentially have a role in treating human AITD.


Asunto(s)
Alcaloides/farmacología , Presentación de Antígeno/efectos de los fármacos , Cadenas HLA-DRB1/inmunología , Tiroiditis Autoinmune/inmunología , Alcaloides/química , Animales , Cadenas HLA-DRB1/genética , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Transgénicos , Péptidos/genética , Péptidos/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Tiroglobulina/genética , Tiroglobulina/inmunología , Tiroiditis Autoinmune/genética , Tiroiditis Autoinmune/patología
4.
Clin Immunol ; 178: 10-19, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-26057130

RESUMEN

Rheumatoid arthritis (RA) occurs two times more often in women than men. B cell depletion has been shown to be efficacious in treating RA. Our previous studies suggested that antigen presentation via B cells results in a sex-specific immune response in DR4 and DR4/DQ8 mice. Here we evaluated the mechanism of efficacy of the B cell depletion in treating arthritis-susceptible DQ8 mice. The data show that arthritic DQ8 mice treated with anti-CD20 antibody in therapeutic protocols show milder disease severity in females as compared to males, which is associated with decreased antibodies to citrullinated proteins and reduced levels of IL-23 and CCL5. Treatment led to significantly increased numbers of T regulatory and monocyte-derived suppressor F4/80+Gr1hi cells in females as compared to male DQ8 mice. Our observations suggest that therapeutic strategies that target B cells may benefit females while functions of DCs might be relatively more important for men than women.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Linfocitos B/inmunología , Quimiocina CCL5/inmunología , Interleucina-23/inmunología , Células Supresoras de Origen Mieloide/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antirreumáticos/farmacología , Artritis Experimental/genética , Artritis Reumatoide/genética , Linfocitos B/efectos de los fármacos , Antígenos CD28/inmunología , Antígenos CD40/inmunología , Proliferación Celular , Quimiocina CCL5/efectos de los fármacos , Quimiocinas/efectos de los fármacos , Quimiocinas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Citometría de Flujo , Cadenas beta de HLA-DQ/genética , Humanos , Interleucina-23/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Rituximab/farmacología , Caracteres Sexuales , Linfocitos T Reguladores/efectos de los fármacos
5.
J Immunol ; 195(10): 4660-7, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26475924

RESUMEN

MHC, especially HLA-DR3 and HLA-DR2, is one of the most important genetic susceptibility regions for systemic lupus erythematosus. Human studies to understand the role of specific HLA alleles in disease pathogenesis have been hampered by the presence of strong linkage disequilibrium in this region. To overcome this, we produced transgenic mice expressing HLA-DR3 (DRß1*0301) and devoid of endogenous class II (both I-A and I-E genes, AE(0)) on a lupus-prone NZM2328 background (NZM2328.DR3(+)AE(0)). Both NZM2328 and NZM2328.DR3(+)AE(0) mice developed anti-dsDNA and glomerulonephritis, but anti-dsDNA titers were higher in the latter. Although kidney histological scores were similar in NZM2328 and NZM2328.DR3(+)AE(0) mice (7.2 ± 4.3 and 8.6 ± 5.7, respectively, p = 0.48), the onset of severe proteinuria occurred earlier in NZM2328.DR3(+)AE(0) mice compared with NZM2328 mice (median, 5 and 9 mo respectively, p < 0.001). Periarterial lymphoid aggregates, classic wire loop lesions, and occasional crescents were seen only in kidneys from NZM2328.DR3(+)AE(0) mice. Interestingly, NZM2328.DR3(+)AE(0) mice, but not NZM2328 mice, spontaneously developed anti-Smith (Sm) Abs. The anti-Sm Abs were seen in NZM2328.DR3(+)AE(0) mice that were completely devoid of endogenous class II (AE(-/) (-)) but not in mice homozygous (AE(+/+)) or heterozygous (AE(+/-)) for endogenous MHC class II. It appears that only HLA-DR3 molecules can preferentially select SmD-reactive CD4(+) T cells for generation of the spontaneous anti-Sm immune response. Thus, our mouse model unravels a critical role for HLA-DR3 in generating an autoimmune response to SmD and lupus nephritis in the NZM2328 background.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Glomerulonefritis/inmunología , Antígeno HLA-DR3/inmunología , Nefritis Lúpica/inmunología , Proteínas Nucleares snRNP/inmunología , Animales , Anticuerpos Antinucleares/genética , Linfocitos T CD4-Positivos/inmunología , ADN/inmunología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Glomerulonefritis/genética , Antígeno HLA-DR2/inmunología , Antígeno HLA-DR3/genética , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/genética , Ratones , Ratones Noqueados
6.
Mol Cell Proteomics ; 14(4): 893-904, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25635267

RESUMEN

The cytotoxic T-lymphocyte-mediated killing of virus-infected cells requires previous recognition of short viral antigenic peptides bound to human leukocyte antigen class I molecules that are exposed on the surface of infected cells. The cytotoxic T-lymphocyte response is critical for the clearance of human respiratory syncytial virus infection. In this study, naturally processed viral human leukocyte antigen class I ligands were identified with mass spectrometry analysis of complex human leukocyte antigen-bound peptide pools isolated from large amounts of human respiratory syncytial virus-infected cells. Acute antiviral T-cell response characterization showed that viral transcription determines both the immunoprevalence and immunodominance of the human leukocyte antigen class I response to human respiratory syncytial virus. These findings have clear implications for antiviral vaccine design.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Inmunidad Celular/inmunología , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/inmunología , Transcripción Genética , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/inmunología , Extractos Celulares , Línea Celular , Humanos , Epítopos Inmunodominantes/inmunología , Ligandos , Ratones Transgénicos , Datos de Secuencia Molecular , Péptidos/química , Péptidos/inmunología , Proteoma/metabolismo , Virus Sincitial Respiratorio Humano/química , Linfocitos T/inmunología , Espectrometría de Masas en Tándem
7.
J Neuroinflammation ; 13(1): 293, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27855706

RESUMEN

BACKGROUND: We investigated the role of human HLA class I molecules in persistent central nervous system (CNS) injury versus repair following virus infection of the CNS. METHODS: Human class I A11+ and B27+ transgenic human beta-2 microglobulin positive (Hß2m+) mice of the H-2 b background were generated on a combined class I-deficient (mouse beta-2 microglobulin deficient, ß2m0) and class II-deficient (mouse Aß0) phenotype. Intracranial infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL mice results in acute encephalitis with prominent injury in the hippocampus, striatum, and cortex. RESULTS: Following infection with TMEV, a picornavirus, the Aß0.ß2m0 mice lacking active immune responses died within 18 to 21 days post-infection. These mice showed severe encephalomyelitis due to rapid replication of the viral genome. In contrast, transgenic Hß2m mice with insertion of a single human class I MHC gene in the absence of human or mouse class II survived the acute infection. Both A11+ and B27+ mice significantly controlled virus RNA expression by 45 days and did not develop late-onset spinal cord demyelination. By 45 days post-infection (DPI), B27+ transgenic mice showed almost complete repair of the virus-induced brain injury, but A11+ mice conversely showed persistent severe hippocampal and cortical injury. CONCLUSIONS: The findings support the hypothesis that the expression of a single human class I MHC molecule, independent of persistent virus infection, influences the extent of sub frequent chronic neuronal injury or repair in the absence of a class II MHC immune response.


Asunto(s)
Infecciones por Cardiovirus/patología , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Antígenos de Histocompatibilidad Clase I/metabolismo , Theilovirus/fisiología , Análisis de Varianza , Animales , Anticuerpos/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Antígeno HLA-A11/metabolismo , Antígeno HLA-B27/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
8.
Hepatology ; 62(5): 1536-50, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26185095

RESUMEN

UNLABELLED: Autoimmune hepatitis (AIH) in humans is a severe inflammatory liver disease characterized by interface hepatitis, the presence of circulating autoantibodies, and hyper-gammaglobulinemia. There are two types of AIH, type 1 (AIH-1) and type 2 (AIH-2), characterized by distinct autoimmune serology. Patients with AIH-1 are positive for anti-smooth muscle and/or antinuclear autoantibodies, whereas patients with AIH-2 have anti-liver kidney microsomal type 1 and/or anti-liver cytosol type 1 autoantibodies. Cytochrome P4502D6 is the antigenic target of anti-liver kidney microsomal type 1, and formiminotransferase cyclodeaminase is the antigenic target of anti-liver cytosol type 1. It is known that AIH, both types 1 and 2, is strongly linked to the human leukocyte antigen (HLA) alleles -DR3, -DR4, and -DR7. However, direct evidence of the association of HLA with AIH is lacking. We developed a novel mouse model of AIH using the HLA-DR3 transgenic mouse on the nonobese-diabetic background by immunization of HLA-DR3- and HLA-DR3+ nonobese-diabetic mice with a DNA plasmid, coding for human cytochrome P4502D6/formiminotransferase cyclodeaminase fusion protein. Immunization with cytochrome P4502D6/formiminotransferase cyclodeaminase leads to a sustained elevation of alanine aminotransferase, development of antinuclear autoantibodies and anti-liver kidney microsomal type 1/anti-liver cytosol type 1 autoantibodies, chronic immune cell infiltration, and parenchymal fibrosis on liver histology in HLA-DR3+ mice. Immunized mice also showed an enhanced T helper 1 immune response and paucity of the frequency of regulatory T cells in the liver. Moreover, HLA-DR3+ mice with exacerbated AIH showed reduced diversity and total load of gut bacteria. CONCLUSION: Our humanized animal model has provided a novel experimental tool to further elucidate the pathogenesis of AIH and to evaluate the efficacy and safety of immunoregulatory therapeutic interventions in vivo.


Asunto(s)
Hepatitis Autoinmune/etiología , Intestinos/microbiología , Microbiota , Animales , Autoanticuerpos/inmunología , Secuencia de Bases , Citocromo P-450 CYP2D6/inmunología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Antígeno HLA-DR3/inmunología , Humanos , Inmunización , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Linfocitos T Reguladores/inmunología
9.
J Immunol ; 193(6): 2919-30, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25092888

RESUMEN

Life-threatening infections caused by Staphylococcus aureus, particularly the community-acquired methicillin-resistant strains of S. aureus, continue to pose serious problems. Greater virulence and increased pathogenicity of certain S. aureus strains are attributed to higher prevalence of exotoxins. Of these exotoxins, the superantigens (SAg) are likely most pathogenic because of their ability to rapidly and robustly activate the T cells even in extremely small quantities. Therefore, countering SAg-mediated T cell activation using T regulatory cells (Tregs) might be beneficial in diseases such as toxic shock syndrome (TSS). As the normal numbers of endogenous Tregs in a typical host are insufficient, we hypothesized that increasing the Treg numbers by administration of IL-2/anti-IL-2 Ab immune complexes (IL2C) or by adoptive transfer of ex vivo expanded Tregs might be more effective in countering SAg-mediated immune activation. HLA-DR3 transgenic mice that closely recapitulate human TSS were treated with IL2C to increase endogenous Tregs or received ex vivo expanded Tregs. Subsequently, they were challenged with SAg to induce TSS. Analyses of various parameters reflective of TSS (serum cytokine/chemokine levels, multiple organ pathology, and SAg-induced peripheral T cell expansion) indicated that increasing the Tregs failed to mitigate TSS. On the contrary, serum IFN-γ levels were increased in IL2C-treated mice. Exploration into the reasons behind the lack of protective effect of Tregs revealed IL-17 and IFN-γ-dependent loss of Tregs during TSS. In addition, significant upregulation of glucocorticoid-induced TNFR family-related receptor on conventional T cells during TSS could render them resistant to Treg-mediated suppression, contributing to failure of Treg-mediated immune regulation.


Asunto(s)
Enterotoxinas/inmunología , Choque Séptico/inmunología , Superantígenos/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Traslado Adoptivo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/farmacología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/biosíntesis , Glucocorticoides , Cadenas alfa de HLA-DR/genética , Cadenas alfa de HLA-DR/inmunología , Cadenas beta de HLA-DR/genética , Cadenas beta de HLA-DR/inmunología , Antígeno HLA-DR3/genética , Antígeno HLA-DR3/inmunología , Interferón gamma/sangre , Interferón gamma/inmunología , Interleucina-17/inmunología , Interleucina-2/inmunología , Interleucina-2/farmacología , Activación de Linfocitos/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Ratones Transgénicos , Receptores del Factor de Necrosis Tumoral/biosíntesis , Choque Séptico/microbiología , Infecciones Estafilocócicas/inmunología , Regulación hacia Arriba
10.
J Immunol ; 193(10): 4859-70, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25339670

RESUMEN

Multiple sclerosis is an inflammatory, demyelinating disease of the CNS of presumed autoimmune origin. Of all the genetic factors linked with multiple sclerosis, MHC class II molecules have the strongest association. Generation of HLA class II transgenic (Tg) mice has helped to elucidate the role of HLA class II genes in chronic inflammatory and demyelinating diseases. We have shown that the human HLA-DRB1*0301 gene predisposes to proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE), whereas HLA-DQß1*0601 (DQ6) was resistant. We also showed that the DQ6 molecule protects from EAE in DRB1*0301.DQ6 double-Tg mice by producing anti-inflammatory IFN-γ. HLA-DQß1*0302 (DQ8) Tg mice were also resistant to PLP(91-110)-induced EAE, but production of proinflammatory IL-17 exacerbated disease in DRB1*0301.DQ8 mice. To further confirm the role of IFN-γ in protection, we generated DRB1*0301.DQ8 mice lacking IFN-γ (DRB1*0301.DQ8.IFN-γ(-/-)). Immunization with PLP(91-110) peptide caused atypical EAE in DRB1*0301.DQ8.IFN-γ(-/-) mice characterized by ataxia, spasticity, and dystonia, hallmarks of brain-specific disease. Severe brain-specific inflammation and demyelination in DRB1*0301.DQ8.IFN-γ(-/-) mice with minimal spinal cord pathology further confirmed brain-specific pathology. Atypical EAE in DRB1*0301.DQ8.IFN-γ(-/-) mice was associated with increased encephalitogenicity of CD4 T cells and their ability to produce greater levels of IL-17 and GM-CSF compared with DRB1*0301.DQ8 mice. Further, areas with demyelination showed increased presence of CD68(+) inflammatory cells, suggesting an important role for monocytes/microglia in causing brain pathology. Thus, our study supports a protective role for IFN-γ in the demyelination of brain through downregulation of IL-17/GM-CSF and induction of neuroprotective factors in the brain by monocytes/microglial cells.


Asunto(s)
Encéfalo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Interferón gamma/inmunología , Interleucina-17/inmunología , Microglía/inmunología , Monocitos/inmunología , Animales , Encéfalo/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/inmunología , Cadenas beta de HLA-DQ/genética , Cadenas beta de HLA-DQ/inmunología , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/inmunología , Humanos , Interferón gamma/deficiencia , Interferón gamma/genética , Interleucina-17/genética , Ratones , Ratones Noqueados , Microglía/patología , Monocitos/patología , Proteína Proteolipídica de la Mielina , Transducción de Señal
11.
Eur J Immunol ; 44(11): 3429-38, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25103892

RESUMEN

HLA-DRB1*0401 expression in humans has been associated with a predisposition to developing rheumatoid arthritis (RA) and collagen-induced arthritis (CIA), while HLA-DRB1*0402 is not associated with susceptibility. Here, we determined if mice transgenic (Tg) for human *0401 have a CD4+ T-cell repertoire that predetermines proinflammatory cytokine production. The data show that both *0401 and *0402 Tg mice can produce TH1/TH17 cytokines, although the kinetics of response may be different. However, in the context of antigen-specific responses in a CIA model, *0402 Tg mice generate a TH2 response that may explain their resistance to developing arthritis. In addition, a significant subset of naïve CD4+ T cells from *0402 Tg mice can be activated in polarizing conditions to differentiate into Treg cells that produce IFN-γ. *0401 Tg mice harbor memory CD4+ T cells that differentiate into IL-17(+) cells in various polarizing conditions. Our data suggest that *0401 Tg mice generate a strong immune response to lipopolysaccharide and may be efficient in clearing infection, and may *0401 have been evolutionarily selected for this ability. Autoimmunity, such as RA, could likely be a bystander effect of the cytokine storm that, along with the presence of low Treg-cell numbers in *0401 Tg mice, causes immune dysregulation.


Asunto(s)
Artritis Experimental/inmunología , Cadenas HLA-DRB1/inmunología , Linfocitos T Reguladores/citología , Animales , Artritis Reumatoide/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Femenino , Cadenas HLA-DRB1/genética , Inflamación/inmunología , Interleucina-17/biosíntesis , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Transgénicos , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología
12.
J Immunol ; 190(2): 513-8, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23293357

RESUMEN

The MHC in humans encodes the most polymorphic genes, the HLA genes, which are critical for the immune system to clear infection. This can be attributed to strong selection pressure as populations moved to different parts of the world and encountered new kinds of infections, leading to new HLA class II alleles. HLA genes also have the highest relative risk for autoimmune diseases. Three haplotypes, that is, HLA-DR2DQ6, DR4DQ8, and DR3DQ2, account for HLA association with most autoimmune diseases. We hypothesize that these haplotypes, along with their multiple subtypes, have survived bottlenecks of infectious episodes in human history because of their ability to present pathogenic peptides to activate T cells that secrete cytokines to clear infections. Unfortunately, they also present self-peptides/mimics to activate autoreactive T cells secreting proinflammatory cytokines that cause autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Antígenos HLA-D/genética , Antígenos HLA-D/inmunología , Animales , Enfermedades Autoinmunes/prevención & control , Autoinmunidad/genética , Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Susceptibilidad a Enfermedades/inmunología , Haplotipos , Humanos , Subgrupos de Linfocitos T
13.
Infect Immun ; 82(1): 286-97, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166949

RESUMEN

Unlike human malaria parasites that induce persistent infection, some rodent malaria parasites, like Plasmodium yoelii strain 17XNL (Py17XNL), induce a transient (self-curing) malaria infection. Cooperation between CD4 T cells and B cells to produce antibodies is thought to be critical for clearance of Py17XNL parasites from the blood, with major histocompatibility complex (MHC) class II molecules being required for activation of CD4 T cells. In order to better understand the correspondence between murine malaria models and human malaria, and in particular the role of MHC (HLA) class II molecules, we studied the ability of humanized mice expressing human HLA class II molecules to clear Py17XNL infection. We showed that humanized mice expressing HLA-DR4 (DR0401) molecules and lacking mouse MHC class II molecules (EA(0)) have impaired production of specific antibodies to Py17XNL and cannot cure the infection. In contrast, mice expressing HLA-DR4 (DR0402), HLA-DQ6 (DQ0601), HLA-DQ8 (DQ0302), or HLA-DR3 (DR0301) molecules in an EA(0) background were able to elicit specific antibodies and self-cure the infection. In a series of experiments, we determined that the inability of humanized DR0401.EA(0) mice to elicit specific antibodies was due to expansion and activation of regulatory CD4(+) Foxp3(+) T cells (Tregs) that suppressed B cells to secrete antibodies through cell-cell interactions. Treg depletion allowed the DR0401.EA(0) mice to elicit specific antibodies and self-cure the infection. Our results demonstrated a differential role of MHC (HLA) class II molecules in supporting antibody responses to Py17XNL malaria and revealed a new mechanism by which malaria parasites stimulate B cell-suppressogenic Tregs that prevent clearance of infection.


Asunto(s)
Linfocitos B/inmunología , Factores de Transcripción Forkhead/metabolismo , Antígenos HLA-DR/inmunología , Malaria/inmunología , Plasmodium yoelii/inmunología , Linfocitos T Reguladores/inmunología , Análisis de Varianza , Animales , Antígenos HLA-DQ/inmunología , Antígeno HLA-DR3/inmunología , Antígeno HLA-DR4/inmunología , Inmunidad Celular/inmunología , Inmunización , Ratones , Ratones Transgénicos , Linfocitos T Reguladores/citología
14.
Clin Immunol ; 152(1-2): 25-35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24631425

RESUMEN

Individuals carrying DRB1*0401 who smoke cigarettes are at an increased risk of developing severe seropositive RA. To determine how cigarette smoke (CS) interacts with host genetic factors in the induction of RA-associated autoimmunity, we used transgenic mice carrying the RA-susceptible HLA genes DR4 and DQ8, but lacking all endogenous murine class II molecules. Cigarette smoke exposure augmented peptidylarginine deiminase (PAD) enzyme expression, and enhanced immune responses to citrullinated collagen and vimentin. Here we show for the first time that DQ molecules can present citrullinated peptides much more efficiently than native peptides. Interestingly, CS exposure suppressed collagen-induced arthritis (CIA) in DRB1*0401 mice although innate immune response was enhanced. On the other hand, CS exposure exacerbated CIA in DQ8 mice, which was accompanied by an increased expression of Th17 gene transcripts in lungs. These observations suggest that cigarette smoke promotes antigen-specific autoimmunity that is profoundly influenced by host genetic factors.


Asunto(s)
Artritis Reumatoide/inmunología , Antígenos HLA-DQ/genética , Antígeno HLA-DR4/genética , Humo/efectos adversos , Fumar/efectos adversos , Animales , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Reumatoide/genética , Autoinmunidad/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno/inmunología , Citocinas/sangre , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Predisposición Genética a la Enfermedad , Antígenos HLA-DQ/inmunología , Antígeno HLA-DR4/inmunología , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/inmunología , Hidrolasas/metabolismo , Pulmón/citología , Pulmón/inmunología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Desiminasas de la Arginina Proteica , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Vimentina/inmunología
15.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G302-12, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24924747

RESUMEN

Celiac disease (CD) is an enteropathy caused by the ingestion of wheat gluten in genetically susceptible individuals. A complete understanding of the pathogenic mechanisms in CD has been hindered because of the lack of adequate in vivo models. In the present study, we explored the events after the intragastric administration of gliadin and of the albumin/globulin fraction from wheat in human leukocyte antigen-DQ8 transgenic mice (DQ8 mice) treated with indomethacin, an inhibitor of cyclooxygenases (COXs). After 10 days of treatment, mice showed a significant reduction of villus height, increased crypt depth, increased number of lamina propria-activated macrophages, and high basal interferon-γ secretion in mesenteric lymph nodes, all of which were specifically related to gliadin intake, whereas the albumin/globulin fraction of wheat was unable to induce similar changes. Cotreatment with NS-398, a specific inhibitor of COX-2, also induced the intestinal lesion. Enteropathy onset was further characterized by high levels of oxidative stress markers, similar to CD. Biochemical assessment of the small intestine revealed the specific activation of matrix metalloproteinases 2 and 9, high caspase-3 activity, and a significant increase of tissue transglutaminase protein levels associated with the intestinal lesion. Notably, after 30 days of treatment, enteropathic mice developed serum antibodies toward gliadin (IgA) and tissue transglutaminase (IgG). We concluded that gliadin intake in combination with COX inhibition caused a basal inflammatory status and an oxidative stress condition in the small intestine of DQ8 mice, thus triggering the mucosal lesion and, subsequently, an antigen-specific immunity.


Asunto(s)
Enfermedad Celíaca/inducido químicamente , Inhibidores de la Ciclooxigenasa 2/toxicidad , Ciclooxigenasa 2/metabolismo , Gliadina , Antígenos HLA-DQ/metabolismo , Indometacina/toxicidad , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Animales , Apoptosis , Caspasa 3/metabolismo , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/patología , Modelos Animales de Enfermedad , Proteínas de Unión al GTP/inmunología , Proteínas de Unión al GTP/metabolismo , Gliadina/inmunología , Antígenos HLA-DQ/genética , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Nitrobencenos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Sulfonamidas/toxicidad , Factores de Tiempo , Transglutaminasas/inmunología , Transglutaminasas/metabolismo
16.
PLoS Pathog ; 8(2): e1002541, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22383876

RESUMEN

Structural diversity in the peptide binding sites of the redundant classical MHC antigen presenting molecules is strongly selected in humans and mice. Although the encoded antigen presenting molecules overlap in antigen presenting function, differences in polymorphism at the MHC I A, B and C loci in humans and higher primates indicate these loci are not functionally equivalent. The structural basis of these differences is not known. We hypothesize that classical class I loci differ in their ability to direct effective immunity against intracellular pathogens. Using a picornavirus infection model and chimeric H-2 transgenes, we examined locus specific functional determinants distinguishing the ability of class I sister genes to direct effective anti viral immunity. Whereas, parental FVB and transgenic FVB mice expressing the H-2K(b) gene are highly susceptible to persisting Theiler's virus infection within the CNS and subsequent demyelination, mice expressing the D(b) transgene clear the virus and are protected from demyelination. Remarkably, animals expressing a chimeric transgene, comprised primarily of K(b) but encoding the peptide binding domain of D(b), develop a robust anti viral CTL response yet fail to clear virus and develop significant demyelination. Differences in expression of the chimeric K(b)α1α2D(b) gene (low) and D(b) (high) in the CNS of infected mice mirror expression levels of their endogenous H-2(q) counterparts in FVB mice. These findings demonstrate that locus specific elements other than those specifying peptide binding and T cell receptor interaction can determine ability to clear virus infection. This finding provides a basis for understanding locus-specific differences in MHC polymorphism, characterized best in human populations.


Asunto(s)
Genes MHC Clase I/fisiología , Sitios Genéticos/fisiología , Inmunidad Innata/genética , Virus/inmunología , Animales , Eficiencia , Antígenos H-2/química , Antígenos H-2/genética , Antígenos H-2/metabolismo , Células HEK293 , Antígeno de Histocompatibilidad H-2D , Humanos , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Modelos Moleculares , Virosis/genética , Virosis/inmunología
17.
Nature ; 456(7221): 534-8, 2008 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19037317

RESUMEN

Major histocompatibility complex (MHC) class II alleles HLA-DQ8 and the mouse homologue I-A(g7) lacking a canonical aspartic acid residue at position beta57 are associated with coeliac disease and type I diabetes. However, the role of this single polymorphism in disease initiation and progression remains poorly understood. The lack of Asp 57 creates a positively charged P9 pocket, which confers a preference for negatively charged peptides. Gluten lacks such peptides, but tissue transglutaminase (TG2) introduces negatively charged residues at defined positions into gluten T-cell epitopes by deamidating specific glutamine residues on the basis of their spacing to proline residues. The commonly accepted model, proposing that HLA-DQ8 simply favours binding of negatively charged peptides, does not take into account the fact that TG2 requires inflammation for activation and that T-cell responses against native gluten peptides are found, particularly in children. Here we show that beta57 polymorphism promotes the recruitment of T-cell receptors bearing a negative signature charge in the complementary determining region 3beta (CDR3beta) during the response against native gluten peptides presented by HLA-DQ8 in coeliac disease. These T cells showed a crossreactive and heteroclitic (stronger) response to deamidated gluten peptides. Furthermore, gluten peptide deamidation extended the T-cell-receptor repertoire by relieving the requirement for a charged residue in CDR3beta. Thus, the lack of a negative charge at position beta57 in MHC class II was met by negatively charged residues in the T-cell receptor or in the peptide, the combination of which might explain the role of HLA-DQ8 in amplifying the T-cell response against dietary gluten.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Glútenes/inmunología , Antígenos HLA-DQ/genética , Polimorfismo Genético/genética , Amidas/química , Animales , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/inmunología , Reacciones Cruzadas , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Gliadina/química , Gliadina/inmunología , Glútenes/química , Antígenos HLA-DQ/química , Antígenos HLA-DQ/inmunología , Humanos , Hibridomas/inmunología , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/inmunología , Electricidad Estática
18.
Mediators Inflamm ; 2014: 468285, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25024509

RESUMEN

Staphylococcus aureus is capable of causing a spectrum of human illnesses. During serious S. aureus infections, the staphylococcal pathogen-associated molecular patterns (PAMPs) such as peptidoglycan, lipoteichoic acid, and lipoproteins and even intact S. aureus, are believed to act in conjunction with the staphylococcal superantigens (SSAg) to activate the innate and adaptive immune system, respectively, and cause immunopathology. However, recent studies have shown that staphylococcal PAMPs could suppress inflammation by several mechanisms and protect from staphylococcal toxic shock syndrome, a life-threatening systemic disease caused by toxigenic S. aureus. Given the contradictory pro- and anti-inflammatory roles of staphylococcal PAMPs, we examined the effects of S. aureus-derived molecular patterns on immune responses driven by SSAg in vivo using HLA-DR3 and HLA-DQ8 transgenic mice. Our study showed that neither S. aureus-derived peptidoglycans (PGN), lipoteichoic acid (LTA), nor heat-killed Staphylococcus aureus (HKSA) inhibited SSAg-induced T cell proliferation in vitro. They failed to antagonize the immunostimulatory effects of SSAg in vivo as determined by their inability to attenuate systemic cytokine/chemokine response and reduce SSAg-induced T cell expansion. These staphylococcal PAMPs also failed to protect HLA-DR3 as well as HLA-DQ8 transgenic mice from either SSAg-induced toxic shock or pneumonia induced by a SSAg-producing strain of S. aureus.


Asunto(s)
Neumonía/inmunología , Neumonía/metabolismo , Choque Séptico/inmunología , Choque Séptico/metabolismo , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Superantígenos/inmunología , Animales , Antígenos HLA-DQ/genética , Antígeno HLA-DR3/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones Transgénicos , Neumonía/inducido químicamente , Choque Séptico/inducido químicamente
19.
Immunol Rev ; 233(1): 62-78, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20192993

RESUMEN

Predisposition to develop rheumatoid arthritis (RA) has been associated with certain human leukocyte antigen (HLA) class II molecules, although the mechanism is still unknown. Various experimental animal models of inflammatory arthritis have been studied to address the role of major histocompatibility complex (MHC) genes in pathogenesis. We have generated transgenic mice expressing HLA class II molecules (DR and DQ) lacking complete endogenous class II molecules to study the interactions involved between class II molecules (DQ and DR) and to define the immunologic mechanisms in inflammatory arthritis. The HLA transgene can positively select CD4(+) T cells expressing various V beta T-cell receptors, and a peripheral tolerance is maintained to transgenic HLA molecules. The expression of HLA molecules on various cells in these mice is similar to that known in humans. In this review, we describe collagen-induced arthritis as a model for human inflammatory arthritis using these transgenic mice. The transgenic mice carrying RA-susceptible haplotype develop gender-biased inflammatory arthritis with clinical and histopathological similarities to RA. Our studies show that polymorphism of HLA class II genes determine the predisposition to rheumatoid/inflammatory arthritis and the epistatic interactions between HLA-DQ and HLA-DR molecules dictate the severity, progression, and modulation of the disease.


Asunto(s)
Artritis Experimental/genética , Artritis Reumatoide/genética , Antígenos HLA-DQ/genética , Antígenos HLA-DR/genética , Animales , Artritis Experimental/inmunología , Artritis Experimental/prevención & control , Artritis Reumatoide/inmunología , Artritis Reumatoide/prevención & control , Autoanticuerpos/sangre , Linfocitos B/inmunología , Citrulina/inmunología , Colágeno/inmunología , Epítopos , Femenino , Predisposición Genética a la Enfermedad , Antígenos HLA-DQ/inmunología , Antígenos HLA-DR/inmunología , Humanos , Masculino , Ratones , Ratones Transgénicos , Procesamiento Proteico-Postraduccional , Medición de Riesgo , Factores de Riesgo , Factores Sexuales , Linfocitos T/inmunología
20.
Gastroenterology ; 142(2): 316-25.e1-12, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22079593

RESUMEN

BACKGROUND & AIMS: Copolymers of hydroxyethyl methacrylate and styrene sulfonate complex with isolated gliadin (the toxic fraction of gluten) and prevent damage to the intestinal barrier in HLA-HCD4/DQ8 mice. We studied the activity toward gluten and hordein digestion and biologic effects of poly(hydroxyethyl methacrylate-co-styrene sulfonate (P(HEMA-co-SS)). We also investigated the effect of gliadin complex formation in intestinal biopsy specimens from patients with celiac disease. METHODS: We studied the ability of P(HEMA-co-SS) to reduce digestion of wheat gluten and barley hordein into immunotoxic peptides using liquid chromatography-mass spectrometry. The biodistribution and pharmacokinetic profile of orally administered P(HEMA-co-SS) was established in rodents using tritium-labeled polymer. We assessed the capacity of P(HEMA-co-SS) to prevent the immunologic and intestinal effects induced by a gluten-food mixture in gluten-sensitized HLA-HCD4/DQ8 mice after short-term and long-term administration. We measured the effects of gliadin complex formation on cytokine release ex vivo using intestinal biopsy specimens from patients with celiac disease. RESULTS: P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein in vitro, thereby decreasing formation of toxic peptides associated with celiac disease. After oral administration to rodents, P(HEMA-co-SS) was predominantly excreted in feces, even in the presence of low-grade mucosal inflammation and increased intestinal permeability. In gluten-sensitized mice, P(HEMA-co-SS) reduced paracellular permeability, normalized anti-gliadin immunoglobulin A in intestinal washes, and modulated the systemic immune response to gluten in a food mixture. Furthermore, incubation of P(HEMA-co-SS) with mucosal biopsy specimens from patients with celiac disease showed that secretion of tumor necrosis factor-α was reduced in the presence of partially digested gliadin. CONCLUSIONS: The copolymer P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein and attenuated the immune response to gluten in a food mixture in rodents. It might be developed to prevent or reduce gluten-induced disorders in humans.


Asunto(s)
Enfermedad Celíaca/metabolismo , Digestión/efectos de los fármacos , Glútenes/metabolismo , Mucosa Intestinal/efectos de los fármacos , Polihidroxietil Metacrilato/análogos & derivados , Poliestirenos/farmacología , Estirenos/farmacología , Animales , Enfermedad Celíaca/tratamiento farmacológico , Enfermedad Celíaca/inmunología , Cromatografía Liquida , Femenino , Gliadina/metabolismo , Gliadina/toxicidad , Glútenes/toxicidad , Humanos , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Yeyuno/efectos de los fármacos , Yeyuno/inmunología , Yeyuno/patología , Masculino , Espectrometría de Masas , Ratones , Permeabilidad/efectos de los fármacos , Polihidroxietil Metacrilato/farmacocinética , Polihidroxietil Metacrilato/farmacología , Polihidroxietil Metacrilato/uso terapéutico , Poliestirenos/farmacocinética , Poliestirenos/uso terapéutico , Unión Proteica , Distribución Aleatoria , Ratas , Estirenos/farmacocinética , Estirenos/uso terapéutico
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda