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










Base de datos
Intervalo de año de publicación
1.
J Immunol ; 211(11): 1656-1668, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37850953

RESUMEN

Regulatory T cells (Tregs) are critical mediators of immune tolerance and play a diametric role in cancer and autoimmunity. Tumor-infiltrating Tregs are often associated with poor prognosis in solid tumors because their enrichment in the tumor microenvironment contributes to immunosuppression. Conversely, dysregulation in the Treg compartment can disrupt self-tolerance, leading to autoimmunity. In the present study, we describe what is, to our knowledge, a novel regulator of Tregs, the GTPase activator regulator of G protein 1 (RGS1), demonstrating that RGS1-deficient human Tregs show downregulation of Treg-associated genes and are less immunosuppressive. These RGS1-deficient Tregs exhibit perturbations to the FOXP3-c-MYC transcriptional axis and downstream metabolic and autophagy programs by shifting their energy demands toward glycolysis and rendering them less autophagic. Taken together, RGS1 may serve as an apical node of Treg function by regulating the FOXP3-c-MYC transcriptional axis, thereby providing a therapeutic rationale for targeting RGS1 for treatment of cancer and autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Neoplasias , Proteínas RGS , Humanos , Linfocitos T Reguladores , Enfermedades Autoinmunes/patología , Autoinmunidad , Neoplasias/patología , Autofagia/genética , Factores de Transcripción Forkhead/metabolismo , Microambiente Tumoral , Proteínas RGS/genética , Proteínas RGS/metabolismo
2.
J Immunother Cancer ; 10(9)2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36096532

RESUMEN

BACKGROUND: Cancer immunotherapy has revolutionized cancer treatment. However, considering the limited success of immunotherapy to only some cancer types and patient cohorts, there is an unmet need for developing new treatments that will result in higher response rates in patients with cancer. Immunoglobulin-like transcript 2 (ILT2), a LILRB family member, is an inhibitory receptor expressed on a variety of immune cells including T cells, natural killer (NK) cells and different myeloid cells. In the tumor microenvironment, binding of class I MHC (in particular HLA-G) to ILT2 on immune cells mediates a strong inhibitory effect, which manifests in inhibition of antitumor cytotoxicity of T and NK cells, and prevention of phagocytosis of the tumor cells by macrophages. METHODS: We describe here the development and characteristics of BND-22, a novel, humanized monoclonal antibody that selectively binds to ILT2 and blocks its interaction with classical MHC I and HLA-G. BND-22 was evaluated for its binding and blocking characteristics as well as its ability to increase the antitumor activity of macrophages, T cells and NK cells in various in vitro, ex vivo and in vivo systems. RESULTS: Collectively, our data suggest that BND-22 enhances activity of both innate and adaptive immune cells, thus generating robust and comprehensive antitumor immunity. In humanized mice models, blocking ILT2 with BND-22 decreased the growth of human tumors, hindered metastatic spread to the lungs, and prolonged survival of the tumor-bearing mice. In addition, BND-22 improved the antitumor immune response of approved therapies such as anti-PD-1 or anti-EGFR antibodies. CONCLUSIONS: BND-22 is a first-in-human ILT2 blocking antibody which has demonstrated efficient antitumor activity in various preclinical models as well as a favorable safety profile. Clinical evaluation of BND-22 as a monotherapy or in combination with other therapeutics is under way in patients with cancer. TRIAL REGISTRATION NUMBER: NCT04717375.


Asunto(s)
Antígenos HLA-G , Neoplasias , Animales , Antígenos HLA-G/metabolismo , Humanos , Inmunoterapia , Células Asesinas Naturales , Ratones , Neoplasias/tratamiento farmacológico , Linfocitos T , Microambiente Tumoral
4.
Nature ; 603(7900): 328-334, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35197632

RESUMEN

Effective antitumour immunity depends on the orchestration of potent T cell responses against malignancies1. Regression of human cancers has been induced by immune checkpoint inhibitors, T cell engagers or chimeric antigen receptor T cell therapies2-4. Although CD8 T cells function as key effectors of these responses, the role of CD4 T cells beyond their helper function has not been defined. Here we demonstrate that a trispecific antibody to HER2, CD3 and CD28 stimulates regression of breast cancers in a humanized mouse model through a mechanism involving CD4-dependent inhibition of tumour cell cycle progression. Although CD8 T cells directly mediated tumour lysis in vitro, CD4 T cells exerted antiproliferative effects by blocking cancer cell cycle progression at G1/S. Furthermore, when T cell subsets were adoptively transferred into a humanized breast cancer tumour mouse model, CD4 T cells alone inhibited HER2+ breast cancer growth in vivo. RNA microarray analysis revealed that CD4 T cells markedly decreased tumour cell cycle progression and proliferation, and also increased pro-inflammatory signalling pathways. Collectively, the trispecific antibody to HER2 induced T cell-dependent tumour regression through direct antitumour and indirect pro-inflammatory/immune effects driven by CD4 T cells.


Asunto(s)
Neoplasias de la Mama , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Femenino , Humanos , Ratones , Receptor ErbB-2/genética
5.
Oncoimmunology ; 9(1): 1811605, 2020 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-33224628

RESUMEN

TGFß is a pleiotropic cytokine that may have both tumor inhibiting and tumor promoting properties, depending on tissue and cellular context. Emerging data support a role for TGFß in suppression of antitumor immunity. Here we show that SAR439459, a pan-TGFß neutralizing antibody, inhibits all active isoforms of human and murine TGFß, blocks TGFß-mediated pSMAD signaling, and TGFß-mediated suppression of T cells and NK cells. In vitro, SAR439459 synergized with anti-PD1 to enhance T cell responsiveness. In syngeneic tumor models, SAR439459 treatment impaired tumor growth, while the combination of SAR439459 with anti-PD-1 resulted in complete tumor regression and a prolonged antitumor immunity. Mechanistically, we found that TGFß inhibition with PD-1 blockade augmented intratumoral CD8+ T cell proliferation, reduced exhaustion, evoked proinflammatory cytokines, and promoted tumor-specific CD8+ T cell responses. Together, these data support the hypothesis that TGFß neutralization using SAR439459 synergizes with PD-1 blockade to promote antitumor immunity and formed the basis for the ongoing clinical investigation of SAR439459 in patients with cancer (NCT03192345).


Asunto(s)
Terapia de Inmunosupresión , Receptor de Muerte Celular Programada 1 , Animales , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Humanos , Tolerancia Inmunológica , Ratones
6.
Proc Natl Acad Sci U S A ; 111(44): 15741-5, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25331893

RESUMEN

There are many transmembrane receptor-like proteins whose ligands have not been identified. A strategy for finding ligands when little is known about their tissue source is to screen each extracellular protein individually expressed in an array format by using a sensitive functional readout. Taking this approach, we have screened a large collection (3,191 proteins) of extracellular proteins for their ability to activate signaling of an orphan receptor, leukocyte tyrosine kinase (LTK). Only two related secreted factors, FAM150A and FAM150B (family with sequence similarity 150 member A and member B), stimulated LTK phosphorylation. FAM150A binds LTK extracellular domain with high affinity (K(D) = 28 pM). FAM150A stimulates LTK phosphorylation in a ligand-dependent manner. This strategy provides an efficient approach for identifying functional ligands for other orphan receptors.


Asunto(s)
Citocinas/metabolismo , Proteoma/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/fisiología , Citocinas/genética , Femenino , Células HEK293 , Humanos , Masculino , Fosforilación/fisiología , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Proteoma/genética , Proteómica , Proteínas Tirosina Quinasas Receptoras/genética
7.
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
8.
Immunity ; 38(3): 489-501, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23521885

RESUMEN

The motheaten mouse has long served as a paradigm for complex autoimmune and inflammatory disease. Null mutations in Ptpn6, which encodes the nonreceptor protein-tyrosine phosphatase Shp1, cause the motheaten phenotype. However, Shp1 regulates multiple signaling pathways in different hematopoietic cell types, so the cellular and molecular mechanism of autoimmunity and inflammation in the motheaten mouse has remained unclear. By using floxed Ptpn6 mice, we dissected the contribution of innate immune cells to the motheaten phenotype. Ptpn6 deletion in neutrophils resulted in cutaneous inflammation, but not autoimmunity, providing an animal model of human neutrophilic dermatoses. By contrast, dendritic cell deletion caused severe autoimmunity, without inflammation. Genetic and biochemical analysis showed that inflammation was caused by enhanced neutrophil integrin signaling through Src-family and Syk kinases, whereas autoimmunity resulted from exaggerated MyD88-dependent signaling in dendritic cells. Our data demonstrate that disruption of distinct Shp1-regulated pathways in different cell types combine to cause motheaten disease.


Asunto(s)
Autoinmunidad/inmunología , Células Dendríticas/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Animales , Autoinmunidad/genética , Antígeno CD11c/genética , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Calgranulina A/genética , Calgranulina A/inmunología , Calgranulina A/metabolismo , Línea Celular Tumoral , Células Cultivadas , Células Dendríticas/metabolismo , Dermis/inmunología , Dermis/metabolismo , Dermis/patología , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Inflamación/genética , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Neutrófilos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Quinasa Syk , Familia-src Quinasas/inmunología , Familia-src Quinasas/metabolismo
9.
J Cell Sci ; 125(Pt 22): 5535-45, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22976304

RESUMEN

Macrophages migrate to sites of insult during normal inflammatory responses. Integrins guide such migration, but the transmission of signals from integrins into the requisite cytoskeletal changes is poorly understood. We have discovered that the hematopoietic adaptor protein Skap2 is necessary for macrophage migration, chemotaxis, global actin reorganization and local actin reorganization upon integrin engagement. Binding of phosphatidylinositol [3,4,5]-triphosphate to the Skap2 pleckstrin-homology (PH) domain, which relieves its conformational auto-inhibition, is critical for this integrin-driven cytoskeletal response. Skap2 enables integrin-induced tyrosyl phosphorylation of Src-family kinases (SFKs), Adap, and Sirpα, establishing their roles as signaling partners in this process. Furthermore, macrophages lacking functional Sirpα unexpectedly have impaired local integrin-induced responses identical to those of Skap2(-/-) macrophages, and Skap2 requires Sirpα for its recruitment to engaged integrins and for coordinating downstream actin rearrangement. By revealing the positive-regulatory role of Sirpα in a Skap2-mediated mechanism connecting integrin engagement with cytoskeletal rearrangement, these data demonstrate that Sirpα is not exclusively immunoinhibitory, and illuminate previously unexplained observations implicating Skap2 and Sirpα in mouse models of inflammatory disease.


Asunto(s)
Citoesqueleto/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/metabolismo , Receptores Inmunológicos/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Bovinos , Quimiotaxis/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Células HEK293 , Humanos , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Modelos Biológicos , Polimerizacion/efectos de los fármacos , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos
10.
Invest Ophthalmol Vis Sci ; 53(10): 6628-37, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22930722

RESUMEN

PURPOSE: To determine if the progression of age-related macular degeneration (AMD) is associated with complement activation in the eye. METHODS: Immunohistochemistry and ELISAs were used to determine the distribution, concentration, and activation of the alternative pathway complement proteases factor B (FB) and factor D (FD) and the central complement protein C3 in genotyped human postmortem donor eyes graded as having no or minimal drusen (category 1; controls), large drusen (category 3), and large drusen with advanced AMD (category 4). RESULTS: C3, FB, and FD were present in vitreous and Bruch's membrane choroid (BM/C) interface of the macula of eyes in all tested AMD severity categories (n = 100). C3, FB, and FD were predominantly located to the choroidal vasculature and Bruch's membrane and, together with the serum proteins transferrin and albumin, elevated in BM/C extracts of category 4 eyes (n = 23) compared with category 1 eyes (n = 24). A significant increase in FB activation was found only in vitreous of category 4 eyes (n = 23) compared with category 1 eyes (n = 25). Genetic variants of complement factor H (CFH), C3, C2, and FB associated with increased risk of AMD were correlated with alternative pathway complement activation in vitreous, but not with complement proteins in BM/C protein extracts. CONCLUSIONS: Increased activation of the alternative complement pathway in vitreous was controlled by disease stage and genetic variation in the complement pathway, supporting a role for complement activation in AMD disease pathogenesis.


Asunto(s)
Complemento C3/metabolismo , Factor B del Complemento/metabolismo , Factor D del Complemento/metabolismo , Vía Alternativa del Complemento/genética , Degeneración Macular/genética , Cuerpo Vítreo/metabolismo , Anciano , Anciano de 80 o más Años , Lámina Basal de la Coroides/metabolismo , Coroides/metabolismo , Activación de Complemento , Complemento C3/genética , Factor B del Complemento/genética , Factor D del Complemento/genética , Factor H de Complemento/genética , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Genotipo , Humanos , Inmunohistoquímica , Degeneración Macular/metabolismo , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Donantes de Tejidos
11.
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
12.
PLoS One ; 6(4): e19233, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559430

RESUMEN

BACKGROUND: MMTV-Cre mouse lines have played important roles in our understanding about the functions of numerous genes in mouse mammary epithelial cells during mammary gland development and tumorigenesis. However, numerous studies have not included MMTV-Cre mice as controls, and many investigators have not indicated which of the different MMTV-Cre founder lines were used in their studies. Here, we describe a lactation defect that severely limits the use of one of the most commonly used MMTV-Cre founder lines. METHODOLOGY/PRINCIPAL FINDINGS: To explore the role of protein tyrosine phosphatase Shp1 in mammary gland development, mice bearing the floxed Shp1 gene were crossed with MMTV-Cre mice and mammary gland development was examined by histological and biochemical techniques, while lactation competency was assessed by monitoring pup growth. Surprisingly, both the Shp1fl/+;MMTV-Cre and MMTV-Cre female mice displayed a severe lactation defect when compared to the Shp1 fl/+ control mice. Histological and biochemical analyses reveal that female mice expressing the MMTV-Cre transgene, either alone or in combination with floxed genes, exhibit defects in lobuloalveolar expansion, presence of large cytoplasmic lipid droplets in luminal alveolar epithelial cells postpartum, and precocious induction of involution. Using a PCR-based genotyping method, the three different founder lines can be distinguished, and we determined that the MMTV-Cre line A, the most widely used MMTV-Cre founder line, exhibits a profound lactation defect that limits its use in studies on mammary gland development. CONCLUSIONS/SIGNIFICANCE: The identification of a lactation defect in the MMTV-Cre line A mice indicates that investigators must use MMTV-Cre alone mice as control in studies that utilize Cre recombinase to excise genes of interest from mammary epithelial cells. Our results also suggest that previous results obtained in studies using the MMTV-Cre line A line should be re-evaluated if the controls did not include mice expressing only Cre recombinase.


Asunto(s)
Lactancia/genética , Glándulas Mamarias Animales/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Animales , Apoptosis , Femenino , Integrasas/metabolismo , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa/métodos , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT5/genética
13.
J Immunol ; 185(6): 3256-67, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20696858

RESUMEN

During responses against viruses and malignancies, naive CD8 T lymphocytes expand to form both short-lived effector cells and a population containing cells with the potential to be long-lived and participate in memory responses (memory precursor effector cells). The strength of antigenic, costimulatory, and cytokine signals during responses impacts the magnitude and type of CD8 populations formed. In vitro studies have revealed that the tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP-1) regulates signal transduction from receptors on T cells including the TCR, helping set the activation threshold, and therefore may shape responses of mature CD8 T cells in vivo. Analysis of CD8 T cells from motheaten mice, which are globally deficient in SHP-1, proved problematic due to cell-extrinsic effects of SHP-1 deficiency in non-T cells on CD8 T cells. Therefore, a conditional knockout of SHP-1 in mature single-positive T cells was developed to analyze cell-intrinsic consequences of complete and partial SHP-1 deficiency on CD8 T cell responses to acute viral infection. The results demonstrated that SHP-1 has disparate effects on subpopulations of responding cells, limiting the magnitude and quality of primary and secondary responses by reducing the number of short-lived effector cells generated without affecting the size of the memory precursor effector cell pool that leads to formation of long-term memory.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Inhibidores de Crecimiento/fisiología , Memoria Inmunológica , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Línea Celular , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Cricetinae , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Inhibidores de Crecimiento/deficiencia , Inhibidores de Crecimiento/genética , Memoria Inmunológica/genética , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Células Madre/citología , Células Madre/inmunología , Células Madre/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
14.
Immunity ; 27(1): 35-48, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17600736

RESUMEN

Spontaneous loss-of-function mutations in the protein-tyrosine phosphatase Shp1 cause the motheaten phenotype, characterized by widespread inflammation and autoimmunity. Because Shp1 is expressed in all hematopoietic cells, it has been unclear which aspects of the motheaten phenotypes are primary effects of Shp1 deficiency. We generated mice (Ptpn6(f/f);CD19-cre) that delete Shp1 specifically in B cells. Analysis of these mice indicates that the increase in B-1a cells in motheaten mice is a cell-autonomous consequence of Shp1 deficiency. Shp1-deficient B-1a cells could be derived from adult bone marrow and had N-nucleotide additions, consistent with an adult origin. Shp1 deficiency altered calcium response evoked by B cell antigen receptors and impaired CD40-evoked proliferation. Young Ptpn6(f/f);CD19-cre mice exhibited elevated serum immunoglobulins and impaired antibody responses to immunization, whereas older Ptpn6(f/f);CD19-cre mice developed systemic autoimmunity, characterized by DNA antibodies and immune complex glomerulonephritis. Thus, Shp1 deficiency in B cells alone perturbs B cell development and causes autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/genética , Subgrupos de Linfocitos B/enzimología , Subgrupos de Linfocitos B/inmunología , Diferenciación Celular/inmunología , Eliminación de Gen , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Enfermedades Autoinmunes/patología , Subgrupos de Linfocitos B/citología , Diferenciación Celular/genética , Células Cultivadas , Glomerulonefritis/enzimología , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia
15.
Annu Rev Immunol ; 25: 473-523, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17291189

RESUMEN

Tyrosyl phosphorylation plays a critical role in multiple signaling pathways regulating innate and acquired immunity. Although tyrosyl phosphorylation is a reversible process, we know much more about the functions of protein-tyrosine kinases (PTKs) than about protein-tyrosine phosphatases (PTPs). Genome sequencing efforts have revealed a large and diverse superfamily of PTPs, which can be subdivided into receptor-like (RPTPs) and nonreceptor (NRPTPs). The role of the RPTP CD45 in immune cell signaling is well known, but those of most other PTPs remain poorly understood. Here, we review the mechanism of action, regulation, and physiological functions of NRPTPs in immune cell signaling. Such an analysis indicates that PTPs are as important as PTKs in regulating the immune system.


Asunto(s)
Genoma Humano/inmunología , Inmunidad Innata , Familia de Multigenes/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Transducción de Señal/inmunología , Animales , Genoma Humano/genética , Humanos , Familia de Multigenes/genética , Proteínas Tirosina Fosfatasas/genética , Transducción de Señal/genética
16.
Immunity ; 25(5): 769-81, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17081782

RESUMEN

T cell activation through the T cell receptor (TCR) is subsequently modified by secondary signals that are either stimulatory or inhibitory. We show that CEACAM1 adhesion molecule isoforms containing a long cytoplasmic domain inhibited multiple T cell functions as a consequence of TCR ligation. Overexpression of CEACAM1 resulted in decreased proliferation, allogeneic reactivity, and cytokine production in vitro and delayed type hypersensitivity and inflammatory bowel disease in mouse models in vivo. Conditioned deletion of CEACAM1 in T cells caused increased TCR-CD3 complex signaling. This T cell regulation was dependent upon the presence of immunoreceptor tyrosine-based inhibition motifs (ITIM) within the cytoplasmic domain of CEACAM1 and the Src homology 2 domain-containing protein tyrosine-phosphatase 1 (SHP1) in the T cell. Thus, CEACAM1 overexpression or deletion in T cells resulted in T cell inhibition or activation, respectively, revealing a role for CEACAM1 as a class of inhibitory receptors potentially amenable to therapeutic manipulation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Antígeno Carcinoembrionario/inmunología , Activación de Linfocitos/inmunología , Isoformas de Proteínas/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Traslado Adoptivo , Secuencias de Aminoácidos , Animales , Linfocitos T CD4-Positivos/metabolismo , Antígeno Carcinoembrionario/química , Colitis/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Immunoblotting , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas/química , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Transfección
17.
J Immunol ; 175(5): 3235-43, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16116214

RESUMEN

Cytotoxic lymphocytes express a large family of granule serine proteases, including one member, granzyme (Grz)M, with a unique protease activity, restricted expression, and distinct gene locus. Although a number of Grzs, including GrzM, have been shown to mediate target cell apoptosis in the presence of perforin, the biological activity of Grz has been restricted to control of a number of viral pathogens, including two natural mouse pathogens, ectromelia, and murine CMV (MCMV). In this article, we describe the first reported gene targeting of GrzM in mice. GrzM-deficient mice display normal NK cell/T cell development and homeostasis and intact NK cell-mediated cytotoxicity of tumor targets as measured by membrane damage and DNA fragmentation. GrzM-deficient mice demonstrated increased susceptibility to MCMV infection typified by the presence of more viral inclusions and transiently higher viral burden in the visceral organs of GrzM-deficient mice compared with wild-type (WT) mice. The cytotoxicity of NK cells from MCMV-infected GrzM-deficient mice remained unchanged and, like WT control mice, GrzM-deficient mice eventually effectively cleared MCMV infection from the visceral organs. In contrast, GrzM-deficient mice were as resistant as WT control mice to mouse pox ectromelia infection, as well as challenge with a number of NK cell-sensitive tumors. These data confirm a role for GrzM in the host response to MCMV infection, but suggest that GrzM is not critical for NK cell-mediated cytotoxicity.


Asunto(s)
Ectromelia Infecciosa/inmunología , Infecciones por Herpesviridae/inmunología , Muromegalovirus , Serina Endopeptidasas/fisiología , Animales , Citotoxicidad Inmunológica , Granzimas , Infecciones por Herpesviridae/patología , Homeostasis , Células Asesinas Naturales/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Muromegalovirus/inmunología , Neoplasias Experimentales/inmunología , Linfocitos T/fisiología
18.
Proc Natl Acad Sci U S A ; 102(29): 10238-43, 2005 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-16006513

RESUMEN

Mice with heterozygous deletion of the PTEN tumor suppressor gene develop a range of epithelial neoplasia as well as lymphoid hyperplasia. Previous studies suggest that PTEN suppresses tumor formation by acting as a phosphoinositide phosphatase to limit signaling by phosphoinositide 3-kinase (PI3K). Here, we examined the effect of deleting various regulatory subunits of PI3K (p85alpha and p85beta) on epithelial neoplasia and lymphoid hyperplasia in PTEN+/- mice. Interestingly, we found the loss of one p85alpha allele with or without the loss of p85beta led to increased incidence of intestinal polyps. Signaling downstream of PI3K was enhanced in the PTEN+/-p85alpha+/-p85beta-/- polyps, as judged by an increased fraction of both cells with cytoplasmic staining of the transcription factor FKHR and cells with positive staining for the proliferation marker Ki-67. In contrast, the incidence of prostate intraepithelial neoplasia was not significantly altered in PTEN+/- mice heterozygous for p85alpha or null for p85beta, whereas the fraction of proliferating cells in prostate intraepithelial neoplasia was reduced in mice lacking p85beta. Finally, there was no significant change in T lymphocyte hyperplasia in the PTEN+/- mice with various p85 deletions, although anti-CD3-stimulated AKT activation was somewhat reduced in the p85alpha+/- background. These results indicate that decreasing the levels of different p85 regulatory subunits can result in enhanced PI3K signaling in some tissues and decreased PI3K signaling in others, supporting the model that, although p85 proteins are essential for class I(A) PI3K signaling, they can function as inhibitors of PI3K signaling in some tissues and thus suppress tumor formation.


Asunto(s)
Enfermedad de Castleman/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Western Blotting , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Heterocigoto , Inmunohistoquímica , Pólipos Intestinales/metabolismo , Antígeno Ki-67 , Masculino , Ratones , Fosfohidrolasa PTEN , Secuencias Reguladoras de Ácidos Nucleicos/genética , Linfocitos T/metabolismo , Factores de Transcripción
19.
Nat Med ; 10(8): 849-57, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15273746

RESUMEN

Noonan syndrome is a common human autosomal dominant birth defect, characterized by short stature, facial abnormalities, heart defects and possibly increased risk of leukemia. Mutations of Ptpn11 (also known as Shp2), which encodes the protein-tyrosine phosphatase Shp2, occur in approximately 50% of individuals with Noonan syndrome, but their molecular, cellular and developmental effects, and the relationship between Noonan syndrome and leukemia, are unclear. We generated mice expressing the Noonan syndrome-associated mutant D61G. When homozygous, the D61G mutant is embryonic lethal, whereas heterozygotes have decreased viability. Surviving Ptpn11(D61G/+) embryos ( approximately 50%) have short stature, craniofacial abnormalities similar to those in Noonan syndrome, and myeloproliferative disease. Severely affected Ptpn11(D61G/+) embryos ( approximately 50%) have multiple cardiac defects similar to those in mice lacking the Ras-GAP protein neurofibromin. Their endocardial cushions have increased Erk activation, but Erk hyperactivation is cell and pathway specific. Our results clarify the relationship between Noonan syndrome and leukemia and show that a single Ptpn11 gain-of-function mutation evokes all major features of Noonan syndrome by acting on multiple developmental lineages in a gene dosage-dependent and pathway-selective manner.


Asunto(s)
Dosificación de Gen , Mutación/genética , Síndrome de Noonan/genética , Proteínas Tirosina Fosfatasas/genética , Animales , Linaje de la Célula/genética , Anomalías Craneofaciales/genética , Modelos Animales de Enfermedad , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/metabolismo , Citometría de Flujo , Genes Dominantes , Cardiopatías Congénitas/genética , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Mutantes , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Trastornos Mieloproliferativos/genética , Pruebas de Precipitina , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/metabolismo
20.
Trends Biochem Sci ; 28(6): 284-93, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12826400

RESUMEN

Src homology-2 (SH2) domain-containing phosphatases (Shps) are a small, highly conserved subfamily of protein-tyrosine phosphatases, members of which are present in both vertebrates and invertebrates. The mechanism of regulation of Shps by ligand binding is now well understood. Much is also known about the normal signaling pathways regulated by each Shp and the consequences of Shp deficiency. Recent studies have identified mutations in human Shp2 as the cause of the inherited disorder Noonan syndrome. Shp2 mutations might also contribute to the pathogenesis of some leukemias. In addition, Shp2 might be a key virulence determinant for the important human pathogen Helicobacter pylori. Despite these efforts, however, the key targets of each Shp have remained elusive. Identifying these substrates remains a major challenge for future research.


Asunto(s)
Proteínas Tirosina Fosfatasas , Transducción de Señal , Tirosina/metabolismo , Dominios Homologos src , Animales , Sitios de Unión , Humanos , Péptidos y Proteínas de Señalización Intracelular , Leucemia/metabolismo , Mutación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas con Dominio SH2 , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...