Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Gut ; 71(1): 89-99, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33563644

RESUMEN

OBJECTIVES: Alterations in the intestinal microbiota are linked with a wide range of autoimmune and inflammatory conditions, including inflammatory bowel diseases (IBD), where pathobionts penetrate the intestinal barrier and promote inflammatory reactions. In patients with IBD, the ability of intestinal macrophages to efficiently clear invading pathogens is compromised resulting in increased bacterial translocation and excessive immune reactions. Here, we investigated how an IBD-associated loss-of-function variant in the protein tyrosine phosphatase non-receptor type 2 (PTPN2) gene, or loss of PTPN2 expression affected the ability of macrophages to respond to invading bacteria. DESIGN: IBD patient-derived macrophages with wild-type (WT) PTPN2 or carrying the IBD-associated PTPN2 SNP, peritoneal macrophages from WT and constitutive PTPN2-knockout mice, as well as mice specifically lacking PTPN2 in macrophages were infected with non-invasive K12 Escherichia coli, the human adherent-invasive E. coli (AIEC) LF82, or a novel mouse AIEC (mAIEC) strain. RESULTS: Loss of PTPN2 severely compromises the ability of macrophages to clear invading bacteria. Specifically, loss of functional PTPN2 promoted pathobiont invasion/uptake into macrophages and intracellular survival/proliferation by three distinct mechanisms: Increased bacterial uptake was mediated by enhanced expression of carcinoembryonic antigen cellular adhesion molecule (CEACAM)1 and CEACAM6 in PTPN2-deficient cells, while reduced bacterial clearance resulted from defects in autophagy coupled with compromised lysosomal acidification. In vivo, mice lacking PTPN2 in macrophages were more susceptible to mAIEC infection and mAIEC-induced disease. CONCLUSIONS: Our findings reveal a tripartite regulatory mechanism by which PTPN2 preserves macrophage antibacterial function, thus crucially contributing to host defence against invading bacteria.


Asunto(s)
Adhesión Bacteriana , Infecciones por Escherichia coli/inmunología , Macrófagos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Animales , Antígenos CD/metabolismo , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Escherichia coli/genética , Escherichia coli/fisiología , Proteínas Ligadas a GPI/metabolismo , Microbioma Gastrointestinal , Predisposición Genética a la Enfermedad , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética
2.
Adv Sci (Weinh) ; 8(13): 2100292, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34258164

RESUMEN

Immune checkpoint blockade (ICB) is an attractive option in cancer therapy, but its efficacy is still less than expected due to the transient and incomplete blocking and the low responsiveness. Herein, an unprecedented programmable unlocking nano-matryoshka-CRISPR system (PUN) targeting programmed cell death ligand 1 (PD-L1) and protein tyrosine phosphatase N2 (PTPN2) is fabricated for permanent and complete and highly responsive immunotherapy. While PUN is inert at normal physiological conditions, enzyme-abundant tumor microenvironment and preternatural intracellular oxidative stress sequentially trigger programmable unlocking of PUN to realize a nano-matryoshka-like release of CRISPR/Cas9. The successful nucleus localization of CRISPR/Cas9 ensures the highly efficient disruption of PD-L1 and PTPN2 to unleash cascade amplified adaptive immune response via revoking the immune checkpoint effect. PD-L1 downregulation in tumor cells not only disrupts PD-1/PD-L1 interaction to attenuate the immunosurveillance evasion but also spurs potent immune T cell responses to enhance adaptive immunity. Synchronously, inhibition of JAK/STAT pathway is relieved by deleting PTPN2, which promotes tumor susceptibility to CD8+ T cells depending on IFN-γ, thus further amplifying adaptive immune responses. Combining these advances together, PUN exhibits optimal antitumor efficiency and long-term immune memory with negligible toxicity, which provides a promising alternative to current ICB therapy.


Asunto(s)
Inmunidad Adaptativa/inmunología , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/inmunología , Terapia de Inmunosupresión/métodos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Inmunidad Adaptativa/genética , Animales , Antígeno B7-H1/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ratones , Nanopartículas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética
3.
J Exp Med ; 218(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33914023

RESUMEN

Tissue-resident memory T cells (TRM cells) are key elements of tissue immunity. Here, we investigated the role of the regulator of T cell receptor and cytokine signaling, Ptpn2, in the formation and function of TRM cells in skin. Ptpn2-deficient CD8+ T cells displayed a marked defect in generating CD69+ CD103+ TRM cells in response to herpes simplex virus type 1 (HSV-1) skin infection. This was accompanied by a reduction in the proportion of KLRG1- memory precursor cells and a transcriptional bias toward terminal differentiation. Of note, forced expression of KLRG1 was sufficient to impede TRM cell formation. Normalizing memory precursor frequencies by transferring equal numbers of KLRG1- cells restored TRM generation, demonstrating that Ptpn2 impacted skin seeding with precursors rather than downstream TRM cell differentiation. Importantly, Ptpn2-deficient TRM cells augmented skin autoimmunity but also afforded superior protection from HSV-1 infection. Our results emphasize that KLRG1 repression is required for optimal TRM cell formation in skin and reveal an important role of Ptpn2 in regulating TRM cell functionality.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Lectinas Tipo C/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Receptores Inmunológicos/inmunología , Animales , Autoinmunidad/inmunología , Femenino , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Ratones , Ratones Endogámicos C57BL , Piel/inmunología
4.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33001862

RESUMEN

Protein tyrosine phosphatase nonreceptor type 2 (PTPN2) recently emerged as a promising cancer immunotherapy target. We set out to investigate the functional role of PTPN2 in the pathogenesis of human colorectal carcinoma (CRC), as its role in immune-silent solid tumors is poorly understood. We demonstrate that in human CRC, increased PTPN2 expression and activity correlated with disease progression and decreased immune responses in tumor tissues. In particular, stage II and III tumors displayed enhanced PTPN2 protein expression in tumor-infiltrating T cells, and increased PTPN2 levels negatively correlated with expression of PD-1, CTLA4, STAT1, and granzyme A. In vivo, T cell- and DC-specific PTPN2 deletion reduced tumor burden in several CRC models by promoting CD44+ effector/memory T cells, as well as CD8+ T cell infiltration and cytotoxicity in the tumor. In direct relevance to CRC treatment, T cell-specific PTPN2 deletion potentiated anti-PD-1 efficacy and induced antitumor memory formation upon tumor rechallenge in vivo. Our data suggest a role for PTPN2 in suppressing antitumor immunity and promoting tumor development in patients with CRC. Our in vivo results identify PTPN2 as a key player in controlling the immunogenicity of CRC, with the strong potential to be exploited for cancer immunotherapy.


Asunto(s)
Neoplasias Colorrectales/inmunología , Proteínas de Neoplasias/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Femenino , Humanos , Memoria Inmunológica , Inmunoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
5.
Front Immunol ; 11: 1856, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973765

RESUMEN

Protein tyrosine phosphatase non-receptor type 2 (PTPN2) plays a pivotal role in immune homeostasis and has been associated with human autoimmune and chronic inflammatory diseases. Though PTPN2 is well-characterized in lymphocytes, little is known about its function in innate immune cells. Our findings demonstrate that dendritic cell (DC)-intrinsic PTPN2 might be the key to explain the central role for PTPN2 in the immune system to maintain immune tolerance. Partial genetic PTPN2 ablation in DCs resulted in spontaneous inflammation, particularly in skin, liver, lung and kidney 22 weeks post-birth. DC-specific PTPN2 controls steady-state immune cell composition and even incomplete PTPN2 deficiency in DCs resulted in enhanced organ infiltration of conventional type 2 DCs, accompanied by expansion of IFNγ-producing effector T-cells. Consequently, the phenotypic effects of DC-specific PTPN2 deficiency were abolished in T-cell deficient Rag knock-out mice. Our data add substantial knowledge about the molecular mechanisms to prevent inflammation and maintain tissue tolerance.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Gut Microbes ; 11(6): 1547-1566, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32586195

RESUMEN

Inflammatory bowel disease (IBD) pathogenesis involves significant contributions from genetic and environmental factors. Loss-of-function single-nucleotide polymorphisms (SNPs) in the protein tyrosine phosphatase non-receptor type 2 (PTPN2) gene increase IBD risk and are associated with altered microbiome population dynamics in IBD. Expansion of intestinal pathobionts, such as adherent-invasive E. coli (AIEC), is strongly implicated in IBD pathogenesis as AIEC increases pro-inflammatory cytokine production and alters tight junction protein regulation - suggesting a potential mechanism of pathogen-induced barrier dysfunction and inflammation. We aimed to determine if PTPN2 deficiency alters intestinal microbiome composition to promote expansion of specific bacteria with pathogenic properties. In mice constitutively lacking Ptpn2, we identified increased abundance of a novel mouse AIEC (mAIEC) that showed similar adherence and invasion of intestinal epithelial cells, but greater survival in macrophages, to the IBD-associated AIEC, LF82. Furthermore, mAIEC caused disease when administered to mice lacking segmented-filamentous bacteria (SFB), and in germ-free mice but only when reconstituted with a microbiome, thus supporting its classification as a pathobiont, not a pathogen. Moreover, mAIEC infection increased the severity of, and prevented recovery from, induced colitis. Although mAIEC genome sequence analysis showed >90% similarity to LF82, mAIEC contained putative virulence genes with >50% difference in gene/protein identities from LF82 indicating potentially distinct genetic features of mAIEC. We show for the first time that an IBD susceptibility gene, PTPN2, modulates the gut microbiome to protect against a novel pathobiont. This study generates new insights into gene-environment-microbiome interactions in IBD and identifies a new model to study AIEC-host interactions.


Asunto(s)
Adhesión Bacteriana , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/inmunología , Escherichia coli/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Animales , Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Femenino , Microbioma Gastrointestinal , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética
7.
Aging (Albany NY) ; 13(2): 2768-2779, 2020 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-33411686

RESUMEN

Atherosclerosis is the main cause of cardiovascular disease. Systemic inflammation is one important characteristic in atherosclerosis. Pro-inflammatory macrophages can secrete inflammatory factors and promote the inflammation of atherosclerosis. It has a great value for the treatment of atherosclerosis by inhibiting the release of inflammatory factors in macrophages. However, the detailed mechanism of this process is still unclear. In this study, we constructed an APOE-/- mice model of atherosclerosis to research the molecular mechanism of atherosclerosis. Protein tyrosine phosphatase non-receptor type 2 (PTPN2), an anti-inflammatory gene, was dramatically decreased in inflammatory mice. Deletion of PTPN2 could significantly induce monocytes toward M1 phenotype of macrophages, enhance the secretion of IL-12 and IL-1, and promote cell proliferation, invasion and metastasis. Mechanism research showed that PTPN2-mediated p65/p38/STAT3 de-phosphorylation could block the process of macrophage inflammation. In vivo experiments showed that PTPN2 may effectively inhibit the inflammatory response during atherosclerosis. In conclusion, we uncovered the negative role of PTPN2 in the occurrence of atherosclerosis, and this study provides a new potential target for atherosclerosis treatment.


Asunto(s)
Aterosclerosis/genética , Proliferación Celular/genética , Inflamación/genética , Macrófagos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Animales , Aterosclerosis/inmunología , Movimiento Celular , Humanos , Inflamación/inmunología , Interleucina-12/genética , Interleucina-1beta/genética , Interleucina-6/genética , Ratones , Ratones Noqueados para ApoE , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células THP-1 , Factor de Transcripción ReIA/inmunología , Factor de Transcripción ReIA/metabolismo , Células U937 , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Diabetes ; 68(6): 1251-1266, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30936146

RESUMEN

Genome-wide association studies have identified PTPN2 as an important non-MHC gene for autoimmunity. Single nucleotide polymorphisms that reduce PTPN2 expression have been linked with the development of various autoimmune disorders, including type 1 diabetes. The tyrosine phosphatase PTPN2 attenuates T-cell receptor and cytokine signaling in T cells to maintain peripheral tolerance, but the extent to which PTPN2 deficiency in T cells might influence type 1 diabetes onset remains unclear. NOD mice develop spontaneous autoimmune type 1 diabetes similar to that seen in humans. In this study, T-cell PTPN2 deficiency in NOD mice markedly accelerated the onset and increased the incidence of type 1 diabetes as well as that of other disorders, including colitis and Sjögren syndrome. Although PTPN2 deficiency in CD8+ T cells alone was able to drive the destruction of pancreatic ß-cells and the onset of diabetes, T-cell-specific PTPN2 deficiency was also accompanied by increased CD4+ T-helper type 1 differentiation and T-follicular-helper cell polarization and increased the abundance of B cells in pancreatic islets as seen in human type 1 diabetes. These findings causally link PTPN2 deficiency in T cells with the development of type 1 diabetes and associated autoimmune comorbidities.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Linfocitos T/metabolismo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Colitis/genética , Colitis/inmunología , Diabetes Mellitus Tipo 1/inmunología , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos NOD , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Síndrome de Sjögren/genética , Síndrome de Sjögren/inmunología , Linfocitos T/inmunología , Células TH1/inmunología , Células TH1/metabolismo
9.
Autoimmunity ; 52(2): 95-101, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-31030572

RESUMEN

OBJECTIVE: To investigate the association between polymorphisms in PTPN2 (rs1893217 and rs478582) and type 1 diabetes (T1D) risk with different diagnosed age, as well as related clinical characteristics in Chinese Han population. METHODS: A total of 2270 Chinese Han individuals (1023 T1D patients and 1247 healthy controls) were genotyped for rs1893217 and rs478582. And 306 newly diagnosed T1D patients were measured for C-peptide levels based on a standard mixed-meal tolerance test. In addition, 40 healthy controls were analyzed for different T cell subsets by multi-color flow cytometry. RESULTS: Neither rs1893217 nor rs478582 showed any association with T1D risk under an additive model. Stratified analysis for T1D diagnosed age revealed that rs1893217, but not rs478582, was significantly associated with T1D patients diagnosed age ≤18 (OR =0.80, 95% CI: 0.67-0.97, p = 0.02). For those diagnosed age >18, neither of them showed any association. We also found that rs1893217 had a higher positive rate of ZnT8A (CC vs. TT carrier, OR = 2.07, 95% CI: 1.07-4.03, p = 0.026) and IA-2A (CT vs. TT carrier, OR = 1.36, 95% CI: 1.02-1.80, p = 0.038). Furthermore, for rs478582, compared with TT, healthy individuals carrying CC/CT carriers had significantly lower frequency and Helios expression of naive Treg subsets (p = 0.049 and 0.048 respectively), but not secreting or activating Treg subsets. In addition, we did not find any association between these two polymorphisms and residual ß-cell function in newly diagnosed T1D patients. CONCLUSIONS: Our results suggest that rs1893217 may increase the risk of early-onset T1D and affect humoral immunity, while rs478582 may affect Treg subsets.


Asunto(s)
Diabetes Mellitus Tipo 1 , Modelos Genéticos , Modelos Inmunológicos , Polimorfismo Genético , Proteína Tirosina Fosfatasa no Receptora Tipo 2 , Linfocitos T Reguladores/inmunología , Adulto , Pueblo Asiatico/etnología , Pueblo Asiatico/genética , China/etnología , Diabetes Mellitus Tipo 1/etnología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología
10.
Nat Commun ; 10(1): 1668, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971695

RESUMEN

Therapies that target the function of immune cells have significant clinical efficacy in diseases such as cancer and autoimmunity. Although functional genomics has accelerated therapeutic target discovery in cancer, its use in primary immune cells is limited because vector delivery is inefficient and can perturb cell states. Here we describe CHIME: CHimeric IMmune Editing, a CRISPR-Cas9 bone marrow delivery system to rapidly evaluate gene function in innate and adaptive immune cells in vivo without ex vivo manipulation of these mature lineages. This approach enables efficient deletion of genes of interest in major immune lineages without altering their development or function. We use this approach to perform an in vivo pooled genetic screen and identify Ptpn2 as a negative regulator of CD8+ T cell-mediated responses to LCMV Clone 13 viral infection. These findings indicate that this genetic platform can enable rapid target discovery through pooled screening in immune cells in vivo.


Asunto(s)
Inmunidad Adaptativa/genética , Sistemas CRISPR-Cas/genética , Técnicas de Transferencia de Gen , Pruebas Genéticas/métodos , Inmunidad Innata/genética , Animales , Trasplante de Médula Ósea , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Estudios de Factibilidad , Femenino , Vectores Genéticos/genética , Genómica/métodos , Células HEK293 , Humanos , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , ARN Guía de Kinetoplastida/genética , Quimera por Trasplante , Células Vero
11.
J Clin Invest ; 129(3): 1193-1210, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30620725

RESUMEN

Genetic variants at the PTPN2 locus, which encodes the tyrosine phosphatase PTPN2, cause reduced gene expression and are linked to rheumatoid arthritis (RA) and other autoimmune diseases. PTPN2 inhibits signaling through the T cell and cytokine receptors, and loss of PTPN2 promotes T cell expansion and CD4- and CD8-driven autoimmunity. However, it remains unknown whether loss of PTPN2 in FoxP3+ regulatory T cells (Tregs) plays a role in autoimmunity. Here we aimed to model human autoimmune-predisposing PTPN2 variants, the presence of which results in a partial loss of PTPN2 expression, in mouse models of RA. We identified that reduced expression of Ptpn2 enhanced the severity of autoimmune arthritis in the T cell-dependent SKG mouse model and demonstrated that this phenotype was mediated through a Treg-intrinsic mechanism. Mechanistically, we found that through dephosphorylation of STAT3, PTPN2 inhibits IL-6-driven pathogenic loss of FoxP3 after Tregs have acquired RORγt expression, at a stage when chromatin accessibility for STAT3-targeted IL-17-associated transcription factors is maximized. We conclude that PTPN2 promotes FoxP3 stability in mouse RORγt+ Tregs and that loss of function of PTPN2 in Tregs contributes to the association between PTPN2 and autoimmunity.


Asunto(s)
Artritis Reumatoide/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Linfocitos T Reguladores/inmunología , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Linfocitos T Reguladores/patología
13.
Biochem Biophys Res Commun ; 486(3): 759-766, 2017 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-28342869

RESUMEN

Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system, and its pathogenesis remains largely unclear. Much attention has been paid to the role of microRNAs (miRs) in regulation of autoimmune disease. Here, we found, for the first time, that miR-448 expression was significantly increased in periphery blood mononuclear cells (PBMC) and cerebrospinal fluid (CSF) of patients with MS, and its expression positively correlated with the disease severity. We further demonstrated that CD4+ T cells, especially the Th17 lineage, were the major source of miR-448 expression. Using gain- and loss-of-function approaches, we further verified that miR-448 could enhance Th17 differentiation, characterized by up-regulated expression levels of IL-17A and RORγt. Interleukin (IL)-1ß as a potent driver of pathogenic Th17 cells was able to strongly induce miR-448 expression in CD4+ T cells through activating NF-κB pathway. Additionally, we identified that miR-448 directly targeted protein tyrosine phosphatase non-receptor type 2 (PTPN2), which has been known as an anti-inflammatory player with capacity to suppress Th17 differentiation. We also observed markedly decreased expression of PTPN2 in PBMC and CSF of MS patients. Our results suggest that miR-448 might promote Th17 differentiation in MS and thus aggravate the disease through inhibiting PTPN2.


Asunto(s)
Interleucina-17/genética , MicroARNs/genética , Esclerosis Múltiple/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Células Th17/patología , Linfocitos B/inmunología , Linfocitos B/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Estudios de Casos y Controles , Diferenciación Celular , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Interleucina-17/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Luciferasas/genética , Luciferasas/metabolismo , MicroARNs/inmunología , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , FN-kappa B/genética , FN-kappa B/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Cultivo Primario de Células , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Inducción de Remisión , Índice de Severidad de la Enfermedad , Transducción de Señal , Células Th17/inmunología
14.
Cytokine ; 82: 52-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26817397

RESUMEN

Lymphoid malignancies are characterized by an accumulation of genetic lesions that act co-operatively to perturb signaling pathways and alter gene expression programs. The Janus kinases (JAK)-signal transducers and activators of transcription (STATs) pathway is one such pathway that is frequently mutated in leukemia and lymphoma. In response to cytokines and growth factors, a cascade of reversible tyrosine phosphorylation events propagates the JAK-STAT pathway from the cell surface to the nucleus. Activated STAT family members then play a fundamental role in establishing the transcriptional landscape of the cell. In leukemia and lymphoma, somatic mutations have been identified in JAK and STAT family members, as well as, negative regulators of the pathway. Most recently, inactivating mutations in the protein tyrosine phosphatase (PTP) genes PTPN1 (PTP1B) and PTPN2 (TC-PTP) were sequenced in B cell lymphoma and T cell acute lymphoblastic leukemia (T-ALL) respectively. The loss of PTP1B and TC-PTP phosphatase activity is associated with an increase in cytokine sensitivity, elevated JAK-STAT signaling, and changes in gene expression. As inactivation mutations in PTPN1 and PTPN2 are restricted to distinct subsets of leukemia and lymphoma, a future challenge will be to identify in which cellular contexts do they contributing to the initiation or maintenance of leukemogenesis or lymphomagenesis. As well, the molecular mechanisms by which PTP1B and TC-PTP loss co-operates with other genetic aberrations will need to be elucidated to design more effective therapeutic strategies.


Asunto(s)
Quinasas Janus , Leucemia , Linfoma , Proteínas de Neoplasias , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 2 , Factores de Transcripción STAT , Transducción de Señal , Humanos , Quinasas Janus/genética , Quinasas Janus/inmunología , Leucemia/genética , Leucemia/inmunología , Leucemia/patología , Linfoma/inmunología , Linfoma/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología
15.
World J Gastroenterol ; 22(3): 1034-44, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26811645

RESUMEN

Genome wide association studies have associated single nucleotide polymorphisms within the gene locus encoding protein tyrosine phosphatase non-receptor type 2 (PTPN2) with the onset of inflammatory bowel disease (IBD) and other inflammatory disorders. Expression of PTPN2 is enhanced in actively inflamed intestinal tissue featuring a marked up-regulation in intestinal epithelial cells. PTPN2 deficient mice suffer from severe intestinal and systemic inflammation and display aberrant innate and adaptive immune responses. In particular, PTPN2 is involved in the regulation of inflammatory signalling cascades, and critical for protecting intestinal epithelial barrier function, regulating innate and adaptive immune responses, and finally for maintaining intestinal homeostasis. On one hand, dysfunction of PTPN2 has drastic effects on innate host defence mechanisms, including increased secretion of pro-inflammatory cytokines, limited autophagosome formation in response to invading pathogens, and disruption of the intestinal epithelial barrier. On the other hand, PTPN2 function is crucial for controlling adaptive immune functions, by regulating T cell proliferation and differentiation as well as maintaining T cell tolerance. In this way, dysfunction of PTPN2 contributes to the manifestation of IBD. The aim of this review is to present an overview of recent findings on the role of PTPN2 in intestinal homeostasis and the impact of dysfunctional PTPN2 on intestinal inflammation.


Asunto(s)
Enfermedades Inflamatorias del Intestino/enzimología , Intestinos/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Inmunidad Adaptativa , Animales , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Inmunidad Innata , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Intestinos/inmunología , Fenotipo , Polimorfismo de Nucleótido Simple , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Factores de Riesgo , Transducción de Señal
16.
J Autoimmun ; 61: 45-53, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26074154

RESUMEN

In addition to the HLA region numerous other gene loci have shown association with type 1 diabetes. How these polymorphisms exert their function has not been comprehensively described, however. We assessed the effect of 39 single nucleotide polymorphisms (SNP) on the development of autoantibody positivity, on progression from autoantibody positivity to clinical disease and on the specificity of the antibody initiating the autoimmune process in 521 autoantibody-positive and 989 control children from a follow-up study starting from birth. Interestingly, PTPN2 rs45450798 gene polymorphism was observed to strongly affect the progression rate of beta-cell destruction after the appearance of humoral beta-cell autoimmunity. Moreover, primary autoantigen dependent associations were also observed as effect of the IKZF4-ERBB3 region on the progression rate of ß-cell destruction was restricted to children with GAD antibodies as their first autoantibody whereas the effect of the INS rs 689 polymorphism was observed among subjects with insulin as the primary autoantigen. In the whole study cohort, INS rs689, PTPN22 rs2476601 and IFIH1 rs1990760 polymorphisms were associated with the appearance of beta-cell autoantibodies. These findings provide new insights into the role of genetic factors implicated in the pathogenesis of type 1 diabetes. The effect of some of the gene variants is restricted to control the initiation of ß-cell autoimmunity whereas others modify the destruction rate of the ß-cells. Furthermore, signs of primary autoantigen-related pathways were detected.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Predisposición Genética a la Enfermedad , Antígenos HLA-DQ/inmunología , Polimorfismo de Nucleótido Simple/inmunología , Alelos , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Estudios de Cohortes , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Diabetes Mellitus Tipo 1/genética , Estudios de Seguimiento , Genotipo , Antígenos HLA-DQ/genética , Humanos , Recién Nacido , Insulina/genética , Insulina/inmunología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Helicasa Inducida por Interferón IFIH1 , Polimorfismo de Nucleótido Simple/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/inmunología , Factores de Riesgo
17.
J Autoimmun ; 53: 105-14, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24997008

RESUMEN

Antigen cross-presentation by dendritic cells is crucial for priming cytotoxic CD8(+) T cells to invading pathogens and tumour antigens, as well as mediating peripheral tolerance to self-antigens. The protein tyrosine phosphatase N2 (PTPN2) attenuates T cell receptor (TCR) signalling and tunes CD8(+) T cell responses in vivo. In this study we have examined the role of PTPN2 in the maintenance of peripheral tolerance after the cross-presentation of pancreatic ß-cell antigens. The transfer of OVA-specific OT-I CD8(+) T cells (C57BL/6) into RIP-mOVA recipients expressing OVA in pancreatic ß-cells only results in islet destruction when OVA-specific CD4(+) T cells are co-transferred. Herein we report that PTPN2-deficient OT-I CD8(+) T cells transferred into RIP-mOVA recipients acquire CTL activity and result in ß cell destruction and the development of diabetes in the absence of CD4(+) help. These studies identify PTPN2 as a critical mediator of peripheral T cell tolerance limiting CD8(+) T cell responses after the cross-presentation of self-antigens. Our findings reveal a mechanism by which PTPN2 SNPs might convert a tolerogenic CD8(+) T cell response into one capable of causing the destruction of pancreatic ß-cells. Moreover, our results provide insight into potential approaches for enhancing T cell-mediated immunity and/or T cell adoptive tumour immunotherapy.


Asunto(s)
Presentación de Antígeno , Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Autoantígenos/genética , Linfocitos T CD4-Positivos/inmunología , Células Secretoras de Insulina/inmunología , Ratones , Ratones Transgénicos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
18.
Nat Immunol ; 15(9): 866-74, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25029551

RESUMEN

The number of Foxp3+ regulatory T cells (Treg cells) must be tightly controlled for efficient suppression of autoimmunity with no impairment of normal immune responses. Here we found that the adaptor TRAF3 was intrinsically required for restraining the lineage determination of thymic Treg cells. T cell-specific deficiency in TRAF3 resulted in a two- to threefold greater frequency of Treg cells, due to the more efficient transition of precursors of Treg cells into Foxp3+ Treg cells. TRAF3 dampened interleukin 2 (IL-2) signaling by facilitating recruitment of the tyrosine phosphatase TCPTP to the IL-2 receptor complex, which resulted in dephosphorylation of the signaling molecules Jak1 and Jak3 and negative regulation of signaling via Jak and the transcription factor STAT5. Our results identify a role for TRAF3 as an important negative regulator of signaling via the IL-2 receptor that affects the development of Treg cells.


Asunto(s)
Diferenciación Celular/inmunología , Interleucina-2/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Timo/citología , Animales , Autoinmunidad/inmunología , Factores de Transcripción Forkhead/inmunología , Janus Quinasa 1/inmunología , Janus Quinasa 3/inmunología , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Factor de Transcripción STAT5/inmunología
19.
Nat Immunol ; 13(5): 439-47, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22513334

RESUMEN

Lymphocyte activation must be tightly regulated to ensure sufficient immunity to pathogens and prevent autoimmunity. Protein tyrosine phosphatases (PTPs) serve critical roles in this regulation by controlling the functions of key receptors and intracellular signaling molecules in lymphocytes. In some cases, PTPs inhibit lymphocyte activation, whereas in others they promote it. Here we discuss recent progress in elucidating the roles and mechanisms of action of PTPs in lymphocyte activation. We also review the accumulating evidence that genetic alterations in PTPs are involved in human autoimmunity.


Asunto(s)
Activación de Linfocitos , Linfocitos/enzimología , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Antígenos CD/inmunología , Autoinmunidad/genética , Autoinmunidad/inmunología , Humanos , Antígenos Comunes de Leucocito/inmunología , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 12/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Superficie Celular/inmunología , Transducción de Señal/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria
20.
Rev Diabet Stud ; 9(4): 188-200, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23804260

RESUMEN

Protein tyrosine phosphatases (PTPs) play a central role in modulating the transduction of cellular signals, including the cells of the immune system. Several PTPs, PTPN22, PTPN2, and UBASH3A, have been associated with risk of type 1 diabetes (T1D) by genome wide association studies. Based on the current understanding of PTPs, it is clear that these variants impact antigen receptor signaling and cytokine signaling. This impact likely contributes to the development and progression of autoimmunity through multiple mechanisms, including failures of central and peripheral tolerance and the promotion of proinflammatory T cell responses. In this review, we discuss the genetic and functional implications of two of these PTPs, PTPN22 and PTPN2, in the development of T1D. We describe the known roles of these proteins in immune function, and how the expression and function of these proteins is altered by the genetic variants associated with T1D. Yet, there are still controversies in the field that require further study and the development of new approaches to extend our understanding of these PTP variants, with the goal of using the information gained to improve our ability to predict and cure T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA