Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 22(3): 381-390, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33589816

RESUMEN

The integrin α4ß7 selectively regulates lymphocyte trafficking and adhesion in the gut and gut-associated lymphoid tissue (GALT). Here, we describe unexpected involvement of the tyrosine phosphatase Shp1 and the B cell lectin CD22 (Siglec-2) in the regulation of α4ß7 surface expression and gut immunity. Shp1 selectively inhibited ß7 endocytosis, enhancing surface α4ß7 display and lymphocyte homing to GALT. In B cells, CD22 associated in a sialic acid-dependent manner with integrin ß7 on the cell surface to target intracellular Shp1 to ß7. Shp1 restrained plasma membrane ß7 phosphorylation and inhibited ß7 endocytosis without affecting ß1 integrin. B cells with reduced Shp1 activity, lacking CD22 or expressing CD22 with mutated Shp1-binding or carbohydrate-binding domains displayed parallel reductions in surface α4ß7 and in homing to GALT. Consistent with the specialized role of α4ß7 in intestinal immunity, CD22 deficiency selectively inhibited intestinal antibody and pathogen responses.


Asunto(s)
Linfocitos B/enzimología , Inmunidad Mucosa , Cadenas beta de Integrinas/metabolismo , Integrinas/metabolismo , Mucosa Intestinal/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/virología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Endocitosis , Femenino , Cadenas beta de Integrinas/inmunología , Integrinas/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/virología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Rotavirus/inmunología , Rotavirus/patogenicidad , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/deficiencia , Lectina 2 Similar a Ig de Unión al Ácido Siálico/genética , Transducción de Señal , Técnicas de Cultivo de Tejidos
2.
Front Immunol ; 11: 576310, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133093

RESUMEN

Shp1, encoded by the gene Ptpn6, is a protein tyrosine phosphatase that transduces inhibitory signals downstream of immunoreceptors in many immune cell types. Blocking Shp1 activity represents an exciting potential immunotherapeutic strategy for the treatment of cancer, as Shp1 inhibition would be predicted to unleash both innate and adaptive immunity against tumor cells. Antibodies blocking the interaction between CD47 on tumor cells and SIRPα on macrophages enhance macrophage phagocytosis, show efficacy in preclinical tumor models, and are being evaluated in the clinic. Here we found that Shp1 bound to phosphorylated peptide sequences derived from SIRPα and transduced the anti-phagocytic signal, as Shp1 loss in mouse bone marrow-derived macrophages increased phagocytosis of tumor cells in vitro. We also generated a novel mouse model to evaluate the impact of global, inducible Ptpn6 deletion on anti-tumor immunity. We found that inducible Shp1 loss drove an inflammatory disease in mice that was phenotypically similar to that seen when Ptpn6 is knocked out from birth. This indicates that acute perturbation of Shp1 in vivo could drive hyperactivation of immune cells, which could be therapeutically beneficial, though at the risk of potential toxicity. In this model, we found that Shp1 loss led to robust anti-tumor immunity against two immune-rich syngeneic tumor models that are moderately inflamed though not responsive to checkpoint inhibitors, MC38 and E0771. Shp1 loss did not promote anti-tumor activity in the non-inflamed B16F10 model. The observed activity in MC38 and E0771 tumors was likely due to effects of both innate and adaptive immune cells. Following Shp1 deletion, we observed increases in intratumoral myeloid cells in both models, which was more striking in E0771 tumors. E0771 tumors also contained an increased ratio of effector to regulatory T cells following Shp1 loss. This was not observed for MC38 tumors, though we did find increased levels of IFNγ, a cytokine produced by effector T cells, in these tumors. Overall, our preclinical data suggested that targeting Shp1 may be an attractive therapeutic strategy for boosting the immune response to cancer via a mechanism involving both innate and adaptive leukocytes.


Asunto(s)
Adenocarcinoma/enzimología , Neoplasias de la Mama/enzimología , Neoplasias del Colon/enzimología , Melanoma Experimental/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Neoplasias Cutáneas/enzimología , Macrófagos Asociados a Tumores/enzimología , Inmunidad Adaptativa , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma/terapia , Animales , Antígenos de Diferenciación/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Femenino , Humanos , Inmunidad Innata , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Receptores Inmunológicos/metabolismo , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Células THP-1 , Carga Tumoral , Microambiente Tumoral , Macrófagos Asociados a Tumores/inmunología
3.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597758

RESUMEN

Gammaherpesviruses are ubiquitous pathogens that establish lifelong infections in the majority of adults worldwide. Chronic gammaherpesvirus infection has been implicated in both lymphomagenesis and, somewhat controversially, autoimmune disease development. Pathogenesis is largely associated with the unique ability of gammaherpesviruses to usurp B cell differentiation, specifically, the germinal center response, to establish long-term latency in memory B cells. The host tyrosine phosphatase SHP1 is known as a brake on immune cell activation and is downregulated in several gammaherpesvirus-driven malignancies. However, here we demonstrate that B cell- but not T cell-intrinsic SHP1 expression supports the gammaherpesvirus-driven germinal center response and the establishment of viral latency. Furthermore, B cell-intrinsic SHP1 deficiency cooperated with gammaherpesvirus infection to increase the levels of double-stranded DNA-reactive antibodies at the peak of viral latency. Thus, in spite of decreased SHP1 levels in gammaherpesvirus-driven B cell lymphomas, B cell-intrinsic SHP1 expression plays a proviral role during the establishment of chronic infection, suggesting that the gammaherpesvirus-SHP1 interaction is more nuanced and is modified by the stage of infection and pathogenesis.IMPORTANCE Gammaherpesviruses establish lifelong infection in a majority of adults worldwide and are associated with a number of malignancies, including B cell lymphomas. These viruses infect naive B cells and manipulate B cell differentiation to achieve a lifelong infection of memory B cells. The germinal center stage of B cell differentiation is important as both an amplifier of the viral latent reservoir and the target of malignant transformation. In this study, we demonstrate that expression of tyrosine phosphatase SHP1, a negative regulator that normally limits the activation and proliferation of hematopoietic cells, enhances the gammaherpesvirus-driven germinal center response and the establishment of chronic infection. The results of this study uncover an intriguing beneficial interaction between gammaherpesviruses that are presumed to profit from B cell activation and a cellular phosphatase that is traditionally perceived to be a negative regulator of the same processes.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Infecciones por Herpesviridae/genética , Interacciones Huésped-Patógeno/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Rhadinovirus/genética , Infecciones Tumorales por Virus/genética , Animales , Anticuerpos Antinucleares/biosíntesis , Linfocitos B/virología , Enfermedad Crónica , ADN/genética , ADN/inmunología , Femenino , Centro Germinal/virología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Memoria Inmunológica , Activación de Linfocitos , Masculino , Ratones , Ratones Transgénicos , Cultivo Primario de Células , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Rhadinovirus/inmunología , Rhadinovirus/patogenicidad , Linfocitos T/inmunología , Linfocitos T/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Latencia del Virus/genética , Latencia del Virus/inmunología
4.
J Neuroimmunol ; 331: 46-57, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29113698

RESUMEN

Glycolysis and mitochondrial respiration are essential for oligodendrocyte metabolism in both the developing and adult CNS. Based on recent reports on the effects of the proinflammatory cytokine IFN-γ on metabolism and on oligodendrocytes, we addressed whether IFN-γ may affect oligodendrocyte bioenergetics in ways relevant to CNS disease. Oligodendrocytes of mice treated with IFN-γ showed significant reductions in aerobic glycolysis and mitochondrial respiration. As expected, IFN-γ treatment led to the induction of STAT1 in oligodendrocytes indicating active signaling into these cells. To determine the direct effects of IFN-γ on oligodendrocyte metabolism, cultured oligodendrocytes were treated with IFN-γ in vitro, which resulted in suppression of glycolysis similar to oligodendrocytes of animals treated with IFN-γ in vivo. Mice lacking SHP-1, a key regulator of IFN-γ and STAT1 signaling in CNS glia, had high constitutive levels of STAT1 and decreased aerobic glycolysis and mitochondrial respiration rates relative to wild type mouse oligodendrocytes. Together, these data show that IFN-γ and SHP-1 control oligodendrocyte bioenergetics in ways that may relate to the role of this cytokine in CNS disease.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Interferón gamma/farmacología , Oligodendroglía/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Animales , Células Cultivadas , Sistema Nervioso Central/patología , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Femenino , Glucólisis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Mutantes Neurológicos , Oligodendroglía/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Factor de Transcripción STAT1/biosíntesis , Factor de Transcripción STAT1/genética , Transducción de Señal/efectos de los fármacos
5.
J Immunol ; 199(1): 129-137, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28550200

RESUMEN

The balance between activation of T cells and their suppression by regulatory T cells (Tregs) is dysregulated in autoimmune diseases and cancer. Autoimmune diseases feature T cells that are resistant to suppression by Tregs, whereas in cancer, T cells are unable to mount antitumor responses due to the Treg-enriched suppressive microenvironment. In this study, we observed that loss of the tyrosine phosphatase SHP-1, a negative regulator of TCR signaling, renders naive CD4+ and CD8+ T cells resistant to Treg-mediated suppression in a T cell-intrinsic manner. At the intracellular level, SHP-1 controlled the extent of Akt activation, which has been linked to the induction of T cell resistance to Treg suppression. Finally, under conditions of homeostatic expansion, SHP-1-deficient CD4+ T cells resisted Treg suppression in vivo. Collectively, these data establish SHP-1 as a critical player in setting the threshold downstream of TCR signaling and identify a novel function of SHP-1 as a regulator of T cell susceptibility to Treg-mediated suppression in vitro and in vivo. Thus, SHP-1 could represent a potential novel immunotherapeutic target to modulate susceptibility of T cells to Treg suppression.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/fisiología , Diferenciación Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Transducción de Señal
6.
Immunol Cell Biol ; 94(8): 802-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27430370

RESUMEN

Adoptive transfer of tumor-specific cytotoxic T cells is a promising advance in cancer therapy. Similarly, checkpoint inhibition has shown striking clinical results in some patients. Here we combine adoptive cell transfer with ablation of the checkpoint protein Src homology 2-domain-containing phosphatase 1 (SHP-1, Ptpn6). Naturally occurring motheaten mice lack SHP-1 and do not survive weaning due to extensive immunopathology. To circumvent this limitation, we created a novel SHP-1(null) mouse that is viable up to 12 weeks of age by knocking out IL1r1. Using this model, we demonstrate that the absence of SHP-1 augments the ability of adoptively transferred CD8(+) T cells to control tumor growth. This therapeutic effect was only observed in situations where T-cell numbers were limited, analogous to clinical settings. However, adoptive transfer of non-CD8(+) SHP-1(null) hematopoietic cells resulted in lethal motheaten-like pathology, indicating that systemic inhibition of SHP-1 could have serious adverse effects. Despite this caveat, our findings support the development of SHP-1 inhibition strategies in human T cells to complement adoptive transfer therapies in the clinic.


Asunto(s)
Traslado Adoptivo , Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Animales , Línea Celular Tumoral , Citometría de Flujo , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/metabolismo
7.
Eur J Immunol ; 46(9): 2103-10, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27354309

RESUMEN

SHP1 is a tyrosine phosphatase critical to proximal regulation of TCR signaling. Here, analysis of CD4-Cre SHP1(fl/fl) conditional knockout thymocytes using CD53, TCRß, CD69, CD4, and CD8α expression demonstrates the importance of SHP1 in the survival of post selection (CD53(+) ), single-positive thymocytes. Using Ca(2+) flux to assess the intensity of TCR signaling demonstrated that SHP1 dampens the signal strength of these same mature, postselection thymocytes. Consistent with its dampening effect, TCR signal strength was also probed functionally using peptides that can mediate selection of the OT-I TCR, to reveal increased negative selection mediated by lower-affinity ligand in the absence of SHP1. Our data show that SHP1 is required for the survival of mature thymocytes and the generation of the functional T-cell repertoire, as its absence leads to a reduction in the numbers of CD4(+) and CD8(+) naïve T cells in the peripheral lymphoid compartments.


Asunto(s)
Selección Clonal Mediada por Antígenos/genética , Selección Clonal Mediada por Antígenos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Timocitos/inmunología , Timocitos/metabolismo , Animales , Biomarcadores , Femenino , Marcación de Gen , Antígenos de Histocompatibilidad/inmunología , Antígenos de Histocompatibilidad/metabolismo , Inmunofenotipificación , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
Int J Mol Med ; 38(1): 45-56, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27221212

RESUMEN

The CD5 transmembrane glycoprotein functions as a co-receptor in the signaling pathway linking T-cell antigen receptor (TCR) engagement to activation and differentiation. Although CD5 effects on TCR signaling have been shown to be primarily inhibitory, the underlying mechanisms remain unclear. In view of recent data revealing the ability of CD5 to associate with the SHP-1 tyrosine phosphatase, a protein that also downregulates TCR signaling, we examined the role of SHP-1 in modulating CD5 function using thymocytes from SHP-1­deficient viable motheaten (mev) mice. The results revealed the association of SHP-1 with CD5 to be markedly increased following TCR stimulation and indicated that this interaction was enhanced by and was dependent on CD5 tyrosine phosphorylation. However, there was no difference of the tyrosine phosphorylation status of CD5 between resting and TCR-stimulated cells in SHP-1­deficient compared to wild-type thymocytes. Lack of SHP-1 activity did not affect the levels of CD5 surface expression, CD5 co-immunoprecipitable tyrosine phosphatase activity and intracellular calcium increase following co-crosslinking of the TCR and CD5. Similarly, an analysis of T­cell thymocyte populations in mev mice expressing an H-Y transgene as well as a construct mediating T­cell restricted CD5 overexpression, revealed that the reduction in the positive selection conferred by CD5 overexpression was unaffected by SHP-1 deficiency. CD5 is not a SHP-1 substrate and SHP-1 is not required for and possibly not involved in the CD5-mediated modulation of TCR signaling.


Asunto(s)
Antígenos CD5/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Membrana Celular/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Timocitos/metabolismo , Timo/metabolismo
9.
Nature ; 521(7552): 357-61, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25799995

RESUMEN

B cells are selected for an intermediate level of B-cell antigen receptor (BCR) signalling strength: attenuation below minimum (for example, non-functional BCR) or hyperactivation above maximum (for example, self-reactive BCR) thresholds of signalling strength causes negative selection. In ∼25% of cases, acute lymphoblastic leukaemia (ALL) cells carry the oncogenic BCR-ABL1 tyrosine kinase (Philadelphia chromosome positive), which mimics constitutively active pre-BCR signalling. Current therapeutic approaches are largely focused on the development of more potent tyrosine kinase inhibitors to suppress oncogenic signalling below a minimum threshold for survival. We tested the hypothesis that targeted hyperactivation--above a maximum threshold--will engage a deletional checkpoint for removal of self-reactive B cells and selectively kill ALL cells. Here we find, by testing various components of proximal pre-BCR signalling in mouse BCR-ABL1 cells, that an incremental increase of Syk tyrosine kinase activity was required and sufficient to induce cell death. Hyperactive Syk was functionally equivalent to acute activation of a self-reactive BCR on ALL cells. Despite oncogenic transformation, this basic mechanism of negative selection was still functional in ALL cells. Unlike normal pre-B cells, patient-derived ALL cells express the inhibitory receptors PECAM1, CD300A and LAIR1 at high levels. Genetic studies revealed that Pecam1, Cd300a and Lair1 are critical to calibrate oncogenic signalling strength through recruitment of the inhibitory phosphatases Ptpn6 (ref. 7) and Inpp5d (ref. 8). Using a novel small-molecule inhibitor of INPP5D (also known as SHIP1), we demonstrated that pharmacological hyperactivation of SYK and engagement of negative B-cell selection represents a promising new strategy to overcome drug resistance in human ALL.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos B/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Transducción de Señal , Secuencias de Aminoácidos/genética , Animales , Antígenos CD/metabolismo , Linfocitos B/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Proteínas de Fusión bcr-abl/genética , Eliminación de Gen , Humanos , Inositol Polifosfato 5-Fosfatasas , Péptidos y Proteínas de Señalización Intracelular/agonistas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Células Precursoras de Linfocitos B/efectos de los fármacos , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B/deficiencia , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasa Syk , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Immunol ; 194(6): 2796-809, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25681345

RESUMEN

Virus-induced myositis is an emerging global affliction that remains poorly characterized with few treatment options. Moreover, muscle-tropic viruses often spread to the CNS, causing dramatically increased morbidity. Therefore, there is an urgent need to explore genetic factors involved in this class of human disease. This report investigates critical innate immune pathways affecting murine virus-induced myositis. Of particular importance, the key immune regulator src homology region 2 domain-containing phosphatase 1 (SHP-1), which normally suppresses macrophage-mediated inflammation, is a major factor in promoting clinical disease in muscle. We show that Theiler's murine encephalomyelitis virus (TMEV) infection of skeletal myofibers induces inflammation and subsequent dystrophic calcification, with loss of ambulation in wild-type (WT) mice. Surprisingly, although similar extensive myofiber infection and inflammation are observed in SHP-1(-/-) mice, these mice neither accumulate dead calcified myofibers nor lose ambulation. Macrophages were the predominant effector cells infiltrating WT and SHP-1(-/-) muscle, and an increased infiltration of immature monocytes/macrophages correlated with an absence of clinical disease in SHP-1(-/-) mice, whereas mature M1-like macrophages corresponded with increased myofiber degeneration in WT mice. Furthermore, blocking SHP-1 activation in WT macrophages blocked virus-induced myofiber degeneration, and pharmacologic ablation of macrophages inhibited muscle calcification in TMEV-infected WT animals. These data suggest that, following TMEV infection of muscle, SHP-1 promotes M1 differentiation of infiltrating macrophages, and these inflammatory macrophages are likely involved in damaging muscle fibers. These findings reveal a pathological role for SHP-1 in promoting inflammatory macrophage differentiation and myofiber damage in virus-infected skeletal muscle, thus identifying SHP-1 and M1 macrophages as essential mediators of virus-induced myopathy.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Diferenciación Celular/inmunología , Macrófagos/inmunología , Miositis/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Theilovirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/virología , Diferenciación Celular/genética , Citometría de Flujo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/virología , Miositis/genética , Miositis/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Theilovirus/fisiología , Transcriptoma/inmunología , Replicación Viral/inmunología
11.
PLoS One ; 9(8): e103685, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25090641

RESUMEN

Allergic rhinitis is a chronic inflammatory disease orchestrated by Th2 lymphocytes. Src homology 2 domain-containing protein tyrosine phosphatase (SHP)-1 is known to be a negative regulator in the IL-4α/STAT-6 signaling pathway of the lung. However, the role of SHP-1 enzyme and its functional relationship with Th2 and Th1 cytokines are not known in the nasal airway. In this study, we aimed to study the nasal inflammation as a result of SHP-1 deficiency in viable motheaten (mev) mice and to investigate the molecular mechanisms involved. Cytology, histology, and expression of cytokines and chemokines were analyzed to define the nature of the nasal inflammation. Targeted gene depletion of Th1 (IFN-γ) and Th2 (IL-4 and IL-13) cytokines was used to identify the critical pathways involved. Matrix metalloproteinases (MMPs) were studied to demonstrate the clearance mechanism of recruited inflammatory cells into the nasal airway. We showed here that mev mice had a spontaneous allergic rhinitis-like inflammation with eosinophilia, mucus metaplasia, up-regulation of Th2 cytokines (IL-4 and IL-13), chemokines (eotaxin), and MMPs. All of these inflammatory mediators were clearly counter-regulated by Th2 and Th1 cytokines. Deletion of IFN-γ gene induced a strong Th2-skewed inflammation with transepithelial migration of the inflammatory cells. These findings suggest that SHP-1 enzyme and Th2/Th1 paradigm may play a critical role in the maintenance of nasal immune homeostasis and in the regulation of allergic rhinitis.


Asunto(s)
Homeostasis , Inflamación/patología , Mucosa Nasal/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Transducción de Señal , Células TH1/inmunología , Células Th2/inmunología , Animales , Quimiocinas/metabolismo , Eosinofilia/inmunología , Eosinofilia/patología , Gelatina , Homeostasis/inmunología , Inflamación/inmunología , Interferón gamma/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Metaplasia , Ratones Endogámicos C57BL , Ratones Mutantes , Moco/metabolismo , Líquido del Lavado Nasal , Mucosa Nasal/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Rinitis/inmunología , Rinitis/patología , Transducción de Señal/inmunología
12.
Arthritis Rheum ; 65(10): 2594-605, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23818173

RESUMEN

OBJECTIVE: Bidirectional interactions between granulocyte-macrophage colony-stimulating factor-positive (GM-CSF+) T cells and interferon regulatory factor 5-positive (IRF-5+) macrophages play a major role in autoimmunity. In the absence of SH2 domain-containing phosphatase 1 (SHP-1), GM-CSF-stimulated cells are resistant to death receptor (DR)-mediated apoptosis. The objective of this study was to determine whether TRA-8, an anti-DR5 agonistic antibody, can eliminate inflammatory macrophages and CD4 T cells in the SHP-1-deficient condition. METHODS: Ubiquitous Cre (Ubc.Cre) human/mouse-chimeric DR5-transgenic mice were crossed with viable SHP-1-defective motheaten (mev/mev) mice. TRA-8 was administered weekly for up to 4 weeks. The clinical scores, histopathologic severity, and macrophage and CD4 T cell phenotypes were evaluated. The role of TRA-8 in depleting inflammatory macrophages and CD4 T cells was also evaluated, using synovial fluid obtained from patients with rheumatoid arthritis (RA). RESULTS: The levels of inflammatory macrophages (interleukin-23-positive [IL-23+] IRF-5+) and CD4 T cells (IL-17+ GM-CSF+) were elevated in mev/mev mice. In DR5-transgenic mev/mev mice, DR5 expression was up-regulated in these 2 cell populations. TRA-8 treatment depleted these cell populations and resulted in a significant reduction in inflammation and in the titers of autoantibodies. In synovial cells from patients with RA, the expression of IRF5 and DR5 was negatively correlated with the expression of PTPN6. TRA-8, but not TRAIL, suppressed RA inflammatory macrophages and Th17 cells under conditions in which the expression of SHP-1 is low. CONCLUSION: In contrast to TRAIL, which lacks the capability to counteract the survival signal in the absence of SHP-1, TRA-8 eliminated both IRF-5+ IL-23+ M1 macrophages and pathogenic GM-CSF+ IL-17+ CD4 T cells in a SHP-1-independent manner. The results of the current study suggest that TRA-8 can deplete inflammatory cell populations that result from a hyperactive GM-CSF/IRF-5 axis.


Asunto(s)
Artritis Reumatoide/patología , Interleucina-23/metabolismo , Macrófagos/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Células TH1/patología , Células Th17/patología , Adulto , Anciano , Animales , Anticuerpos Monoclonales/farmacología , Artritis Reumatoide/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Factores Reguladores del Interferón/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/efectos de los fármacos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Células TH1/metabolismo , Células Th17/metabolismo , Regulación hacia Arriba/efectos de los fármacos
13.
J Exp Med ; 210(7): 1419-31, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23797092

RESUMEN

The protein-tyrosine phosphatase Shp1 is expressed ubiquitously in hematopoietic cells and is generally viewed as a negative regulatory molecule. Mutations in Ptpn6, which encodes Shp1, result in widespread inflammation and premature death, known as the motheaten (me) phenotype. Previous studies identified Shp1 as a negative regulator of TCR signaling, but the severe systemic inflammation in me mice may have confounded our understanding of Shp1 function in T cell biology. To define the T cell­intrinsic role of Shp1, we characterized mice with a T cell­specific Shp1 deletion (Shp1fl/fl CD4-cre). Surprisingly, thymocyte selection and peripheral TCR sensitivity were unaltered in the absence of Shp1. Instead, Shp1(fl/fl) CD4-cre mice had increased frequencies of memory phenotype T cells that expressed elevated levels of CD44. Activation of Shp1-deficient CD4⁺ T cells also resulted in skewing to the Th2 lineage and increased IL-4 production. After IL-4 stimulation of Shp1- deficient T cells, Stat 6 activation was sustained, leading to enhanced Th2 skewing. Accordingly, we observed elevated serum IgE in the steady state. Blocking or genetic deletion of IL-4 in the absence of Shp1 resulted in a marked reduction of the CD44hi population. Therefore, Shp1 is an essential negative regulator of IL-4 signaling in T lymphocytes.


Asunto(s)
Homeostasis/inmunología , Interleucina-4/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Transducción de Señal/inmunología , Linfocitos T/enzimología , Linfocitos T/inmunología , Animales , Memoria Inmunológica , Interleucina-4/deficiencia , Interleucina-4/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Células Th2/enzimología , Células Th2/inmunología
14.
Nature ; 498(7453): 224-7, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23708968

RESUMEN

The protein-tyrosine phosphatase SHP-1 has critical roles in immune signalling, but how mutations in SHP-1 cause inflammatory disease in humans remains poorly defined. Mice homozygous for the Tyr208Asn amino acid substitution in the carboxy terminus of SHP-1 (referred to as Ptpn6(spin) mice) spontaneously develop a severe inflammatory syndrome that resembles neutrophilic dermatosis in humans and is characterized by persistent footpad swelling and suppurative inflammation. Here we report that receptor-interacting protein 1 (RIP1)-regulated interleukin (IL)-1α production by haematopoietic cells critically mediates chronic inflammatory disease in Ptpn6(spin) mice, whereas inflammasome signalling and IL-1ß-mediated events are dispensable. IL-1α was also crucial for exacerbated inflammatory responses and unremitting tissue damage upon footpad microabrasion of Ptpn6(spin) mice. Notably, pharmacological and genetic blockade of the kinase RIP1 protected against wound-induced inflammation and tissue damage in Ptpn6(spin) mice, whereas RIP3 deletion failed to do so. Moreover, RIP1-mediated inflammatory cytokine production was attenuated by NF-κB and ERK inhibition. Together, our results indicate that wound-induced tissue damage and chronic inflammation in Ptpn6(spin) mice are critically dependent on RIP1-mediated IL-1α production, whereas inflammasome signalling and RIP3-mediated necroptosis are dispensable. Thus, we have unravelled a novel inflammatory circuit in which RIP1-mediated IL-1α secretion in response to deregulated SHP-1 activity triggers an inflammatory destructive disease that proceeds independently of inflammasomes and programmed necrosis.


Asunto(s)
Inflamasomas , Interleucina-1alfa/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Muerte Celular , Dermatitis/inmunología , Dermatitis/metabolismo , Dermatitis/patología , Modelos Animales de Enfermedad , Extremidades/patología , Femenino , Eliminación de Gen , Humanos , Inflamasomas/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1alfa/deficiencia , Interleucina-1alfa/genética , Interleucina-1beta/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Transducción de Señal , Cicatrización de Heridas , Heridas y Lesiones/inmunología , Heridas y Lesiones/patología
15.
Cell Death Dis ; 4: e567, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23559001

RESUMEN

Reorganization of spared neural network connections is one of the most important processes for restoring impaired function after brain injury. However, plasticity is quite limited in the adult brain due to the presence of inhibitory molecules and a lack of intrinsic neuronal signals for axonal growth. Src homology 2-containing phosphatase (SHP)-1 has been shown to have a role in axon growth inhibition. Here, we tested the hypothesis that SHP-1 negatively affects axonal reorganization. We observed that unilateral motor cortex injury led to increased expression and activity of SHP-1 in the contralesional cortex. In this model, corticospinal axons originating from the contralesional cortex sprouted into the denervated side of the cervical spinal cord after injury. We observed that the number of sprouting fibers was increased in SHP-1-deficient heterozygous viable motheaten (+/me(v)) mice, which show reduced SHP-1 activity, and in wild-type mice treated with an SHP inhibitor. Motor function recovery of impaired forelimb was enhanced in +/me(v) mice. Collectively, our results indicate that downregulation of SHP-1 activity promotes corticospinal tract sprouting and functional recovery after brain injury.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Corteza Motora/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Tractos Piramidales/efectos de los fármacos , Quinolinas/farmacología , Recuperación de la Función , Animales , Axones/efectos de los fármacos , Axones/fisiología , Lesiones Encefálicas/metabolismo , Regulación hacia Abajo , Miembro Anterior/efectos de los fármacos , Miembro Anterior/fisiología , Heterocigoto , Masculino , Ratones , Corteza Motora/lesiones , Corteza Motora/fisiología , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/fisiología , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Tractos Piramidales/enzimología , Tractos Piramidales/lesiones
16.
J Immunol ; 190(4): 1631-45, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23335748

RESUMEN

Deficiency in Src homology region 2 domain-containing phosphatase 1/protein tyrosine phosphatase nonreceptor type 6 (SHP1/PTPN6) is linked with chronic inflammatory diseases and hematological malignancies in humans. In this study, we exploited the embryonic and larval stages of zebrafish (Danio rerio) as an animal model to study ptpn6 function in the sole context of innate immunity. We show that ptpn6 knockdown induces a spontaneous inflammation-associated phenotype at the late larval stage. Surprisingly, glucocorticoid treatment did not suppress inflammation under ptpn6 knockdown conditions but further enhanced leukocyte infiltration and proinflammatory gene expression. Experiments in a germ-free environment showed that the late larval phenotype was microbe independent. When ptpn6 knockdown embryos were challenged with Salmonella typhimurium or Mycobacterium marinum at earlier stages of development, the innate immune system was hyperactivated to a contraproductive level that impaired the control of these pathogenic bacteria. Transcriptome analysis demonstrated that Kyoto Encyclopedia of Genes and Genomes pathways related to pathogen recognition and cytokine signaling were significantly enriched under these conditions, suggesting that ptpn6 functions as a negative regulator that imposes a tight control over the level of innate immune response activation during infection. In contrast to the hyperinduction of proinflammatory cytokine genes under ptpn6 knockdown conditions, anti-inflammatory il10 expression was not hyperinduced. These results support that ptpn6 has a crucial regulatory function in preventing host-detrimental effects of inflammation and is essential for a successful defense mechanism against invading microbes.


Asunto(s)
Inmunidad Innata/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Infecciones por Salmonella/enzimología , Infecciones por Salmonella/inmunología , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Pez Cebra , Animales , Embrión no Mamífero , Inmunofenotipificación , Modelos Animales , Infecciones por Salmonella/genética
17.
Cancer Res ; 72(22): 5934-44, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22962271

RESUMEN

A genetic screen using a library of 6,961 siRNAs led to the identification of SHP-1 (PTPN6), a tumor suppressor frequently mutated in malignant lymphomas, leukemias, and prostate cancer, as a potential synthetic lethal partner of the DNA repair protein polynucleotide kinase/phosphatase (PNKP). After confirming the partnership with SHP-1, we observed that codepletion of PNKP and SHP-1 induced apoptosis. A T-cell lymphoma cell line that is SHP-1 deficient (Karpas 299) was shown to be sensitive to a chemical inhibitor of PNKP, but resistance was restored by expression of wild-type SHP-1 in these cells. We determined that while SHP-1 depletion does not significantly impact DNA strand-break repair, it does amplify the level of reactive oxygen species (ROS) and elevate endogenous DNA damage. The ROS scavenger WR1065 afforded protection to SHP-1-depleted cells treated with the PNKP inhibitor. We propose that codisruption of SHP-1 and PNKP leads to an increase in DNA damage that escapes repair, resulting in the accumulation of cytotoxic double-strand breaks and induction of apoptosis. This supports an alternative paradigm for synthetic lethal partnerships that could be exploited therapeutically.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Mama/genética , Enzimas Reparadoras del ADN/genética , Neoplasias Pulmonares/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Muerte Celular/genética , Línea Celular Tumoral , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/metabolismo , Femenino , Pruebas Genéticas , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Células MCF-7 , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , ARN Interferente Pequeño/genética , Transfección
18.
J Immunol ; 189(5): 2203-10, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22826316

RESUMEN

The process of phagocytosis and phagosome maturation involves the recruitment of effector proteins that participate in phagosome formation and in the acidification and/or fusion with various endocytic vesicles. In the current study, we investigated the role of the Src homology region 2 domain-containing phosphatase 1 (SHP-1) in phagolysosome biogenesis. To this end, we used immortalized bone marrow macrophages derived from SHP-1-deficient motheaten mice and their wild-type littermates. We found that SHP-1 is recruited early and remains present on phagosomes for up to 4 h postphagocytosis. Using confocal immunofluorescence microscopy and Western blot analyses on purified phagosome extracts, we observed an impaired recruitment of lysosomal-associated membrane protein 1 in SHP-1-deficient macrophages. Moreover, Western blot analyses revealed that whereas the 51-kDa procathepsin D is recruited to phagosomes, it is not processed into the 46-kDa cathepsin D in the absence of SHP-1, suggesting a defect in acidification. Using the lysosomotropic agent LysoTracker as an indicator of phagosomal pH, we obtained evidence that in the absence of SHP-1, phagosome acidification was impaired. Taken together, these results are consistent with a role for SHP-1 in the regulation of signaling or membrane fusion events involved in phagolysosome biogenesis.


Asunto(s)
Células de la Médula Ósea/enzimología , Diferenciación Celular/inmunología , Lisosomas/enzimología , Macrófagos/enzimología , Fagosomas/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Línea Celular Transformada , Femenino , Concentración de Iones de Hidrógeno , Lisosomas/inmunología , Lisosomas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Fagosomas/inmunología , Fagosomas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia
19.
Diabetes ; 61(8): 1949-58, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22698917

RESUMEN

The protein-tyrosine phosphatase Shp1 negatively regulates insulin action on glucose homeostasis in liver and muscle, but its potential role in obesity-linked insulin resistance has not been examined. To investigate the role of Shp1 in hepatic insulin resistance, we generated hepatocyte-specific Shp1 knockout mice (Ptpn6(H-KO)), which were subjected to extensive metabolic monitoring throughout an 8-week standard chow diet (SD) or high-fat diet (HFD) feeding. We report for the first time that Shp1 expression is upregulated in metabolic tissues of HFD-fed obese mice. When compared with their Shp1-expressing Ptpn6(f/f) littermates, Ptpn6(H-KO) mice exhibited significantly lowered fasting glycemia and heightened hepatic insulin sensitivity. After HFD feeding, Ptpn6(H-KO) mice developed comparable levels of obesity as Ptpn6(f/f) mice, but they were remarkably protected from liver insulin resistance, as revealed by euglycemic clamps and hepatic insulin signaling determinations. Although Ptpn6(H-KO) mice still acquired diet-induced peripheral insulin resistance, they were less hyperinsulinemic during a glucose tolerance test because of reduced insulin secretion. Ptpn6(H-KO) mice also exhibited increased insulin clearance in line with enhanced CC1 tyrosine phosphorylation in liver. These results show that hepatocyte Shp1 plays a critical role in the development of hepatic insulin resistance and represents a novel therapeutic target for obesity-linked diabetes.


Asunto(s)
Resistencia a la Insulina/fisiología , Hígado/metabolismo , Obesidad/fisiopatología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Animales , Glucemia/metabolismo , Dieta Alta en Grasa , Eliminación de Gen , Insulina/metabolismo , Ratones , Obesidad/metabolismo
20.
J Immunol ; 188(11): 5397-407, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22539788

RESUMEN

Dendritic cells (DCs) promote immune responses to foreign Ags and immune tolerance to self-Ags. Deregulation of DCs is implicated in autoimmunity, but the molecules that regulate DCs to protect against autoimmunity have remained unknown. In this study, we show that mice lacking the protein tyrosine phosphatase Shp1 specifically in DCs develop splenomegaly associated with more CD11c(+) DCs. Splenic DCs from the mutant mice showed upregulation of CD86 and CCR7 expression and of LPS-induced production of proinflammatory cytokines. The mice manifested more splenic Th1 cells, consistent with the increased ability of their DCs to induce production of IFN-γ by Ag-specific T cells in vitro. The number of splenic CD5(+)CD19(+) B-1a cells and the serum concentrations of Igs M and G2a were also increased in the mutant mice. Moreover, aged mutant mice developed glomerulonephritis and interstitial pneumonitis together with increased serum concentrations of autoantibodies. Shp1 is thus a key regulator of DC functions that protects against autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/genética , Diferenciación Celular/inmunología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Células TH1/inmunología , Animales , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Antígeno CD11c/biosíntesis , Diferenciación Celular/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Esplenomegalia/genética , Esplenomegalia/inmunología , Esplenomegalia/patología , Células TH1/citología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...