Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Immunol ; 211(6): 917-922, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37566514

RESUMO

Ras guanine nucleotide-releasing protein 1 (Rasgrp1) is a Ras guanine nucleotide exchange factor that participates in the activation of the Ras-ERK signaling pathway in developing T cells and is required for efficient thymic T cell positive selection. However, the role of Rasgrp1 in mature peripheral T cells has not been definitively addressed, in part because peripheral T cells from constitutive Rasgrp1-deficient mice show an abnormal activated phenotype. In this study, we generated an inducible Rasgrp1-deficient mouse model to allow acute disruption of Rasgrp1 in peripheral CD4+ T cells in the context of normal T cell development. TCR/CD28-mediated activation of Ras-ERK signaling was blocked in Rasgrp1-deficient peripheral CD4+ T cells. Furthermore, Rasgrp1-deficient CD4+ T cells were unable to synthesize IL-2 and the high-affinity IL-2R and were unable to proliferate in response to TCR/CD28 stimulation. These findings highlight an essential function for Rasgrp1 for TCR/CD28-induced Ras-ERK activation in peripheral CD4+ T cells.


Assuntos
Antígenos CD28 , Linfócitos T CD4-Positivos , Camundongos , Animais , Linfócitos T CD4-Positivos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/metabolismo
2.
Development ; 147(23)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33144395

RESUMO

RASA1, a negative regulator of Ras-MAPK signaling, is essential for the development and maintenance of lymphatic vessel valves. However, whether RASA1 is required for the development and maintenance of lymphovenous valves (LVV) and venous valves (VV) is unknown. In this study, we show that induced disruption of Rasa1 in mouse embryos did not affect initial specification of LVV or central VV, but did affect their continued development. Similarly, a switch to expression of a catalytically inactive form of RASA1 resulted in impaired LVV and VV development. Blocked development of LVV was associated with accumulation of the basement membrane protein, collagen IV, in LVV-forming endothelial cells (EC), and could be partially or completely rescued by MAPK inhibitors and drugs that promote collagen IV folding. Disruption of Rasa1 in adult mice resulted in venous hypertension and impaired VV function that was associated with loss of EC from VV leaflets. In conclusion, RASA1 functions as a negative regulator of Ras signaling in EC that is necessary for EC export of collagen IV, thus permitting the development of LVV and the development and maintenance of VV.


Assuntos
Desenvolvimento Embrionário/genética , Organogênese/genética , Válvulas Venosas/crescimento & desenvolvimento , Proteína p120 Ativadora de GTPase/genética , Animais , Membrana Basal/crescimento & desenvolvimento , Membrana Basal/metabolismo , Colágeno Tipo IV/genética , Embrião de Mamíferos , Células Endoteliais/citologia , Vasos Linfáticos/metabolismo , Camundongos , Válvulas Venosas/metabolismo
3.
J Immunol ; 195(1): 31-5, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26002977

RESUMO

Ras GTPase-activating proteins (RasGAPs) inhibit signal transduction initiated through the Ras small GTP-binding protein. However, which members of the RasGAP family act as negative regulators of T cell responses is not completely understood. In this study, we investigated potential roles for the RasGAPs RASA1 and neurofibromin 1 (NF1) in T cells through the generation and analysis of T cell-specific RASA1 and NF1 double-deficient mice. In contrast to mice lacking either RasGAP alone in T cells, double-deficient mice developed T cell acute lymphoblastic leukemia/lymphoma, which originated at an early point in T cell development and was dependent on activating mutations in the Notch1 gene. These findings highlight RASA1 and NF1 as cotumor suppressors in the T cell lineage.


Assuntos
Neurofibromina 1/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptor Notch1/genética , Proteína p120 Ativadora de GTPase/genética , Animais , Deleção de Genes , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Mutação , Neurofibromina 1/deficiência , Neurofibromina 1/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Receptor Notch1/imunologia , Transdução de Sinais , Baço/imunologia , Baço/patologia , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/patologia , Timo/imunologia , Timo/patologia , Fatores de Tempo , Proteína p120 Ativadora de GTPase/deficiência , Proteína p120 Ativadora de GTPase/imunologia
4.
Blood ; 123(10): 1574-85, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24385536

RESUMO

The hepatic hormone hepcidin is a key regulator of systemic iron metabolism. Its expression is largely regulated by 2 signaling pathways: the "iron-regulated" bone morphogenetic protein (BMP) and the inflammatory JAK-STAT pathways. To obtain broader insights into cellular processes that modulate hepcidin transcription and to provide a resource to identify novel genetic modifiers of systemic iron homeostasis, we designed an RNA interference (RNAi) screen that monitors hepcidin promoter activity after the knockdown of 19 599 genes in hepatocarcinoma cells. Interestingly, many of the putative hepcidin activators play roles in signal transduction, inflammation, or transcription, and affect hepcidin transcription through BMP-responsive elements. Furthermore, our work sheds light on new components of the transcriptional machinery that maintain steady-state levels of hepcidin expression and its responses to the BMP- and interleukin-6-triggered signals. Notably, we discover hepcidin suppression mediated via components of Ras/RAF MAPK and mTOR signaling, linking hepcidin transcriptional control to the pathways that respond to mitogen stimulation and nutrient status. Thus using a combination of RNAi screening, reverse phase protein arrays, and small molecules testing, we identify links between the control of systemic iron homeostasis and critical liver processes such as regeneration, response to injury, carcinogenesis, and nutrient metabolism.


Assuntos
Regulação da Expressão Gênica , Hepcidinas/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Interferência de RNA , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Perfilação da Expressão Gênica , Hepcidinas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ligação Proteica , Reprodutibilidade dos Testes , Elementos de Resposta , Transcrição Gênica
5.
Proc Natl Acad Sci U S A ; 110(21): 8621-6, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23650393

RESUMO

Mutations in gene RASA1 have been historically associated with capillary malformation-arteriovenous malformation, but sporadic reports of lymphatic involvement have yet to be investigated in detail. To investigate the impact of RASA1 mutations in the lymphatic system, we performed investigational near-infrared fluorescence lymphatic imaging and confirmatory radiographic lymphangiography in a Parkes-Weber syndrome (PKWS) patient with suspected RASA1 mutations and correlated the lymphatic abnormalities against that imaged in an inducible Rasa1 knockout mouse. Whole-exome sequencing (WES) analysis and validation by Sanger sequencing of DNA from the patient and unaffected biological parents enabled us to identify an early-frameshift deletion in RASA1 that was shared with the father, who possessed a capillary stain but otherwise no overt disease phenotype. Abnormal lymphatic vasculature was imaged in both affected and unaffected legs of the PKWS subject that transported injected indocyanine green dye to the inguinal lymph node and drained atypically into the abdomen and into dermal lymphocele-like vesicles on the groin. Dermal lymphatic hyperplasia and dilated vessels were observed in Rasa1-deficient mice, with subsequent development of chylous ascites. WES analyses did not identify potential gene modifiers that could explain the variability of penetrance between father and son. Nonetheless, we conclude that the RASA1 mutation is responsible for the aberrant lymphatic architecture and functional abnormalities, as visualized in the PKWS subject and in the animal model. Our unique method to combine investigatory near-infrared fluorescence lymphatic imaging and WES for accurate phenoptyping and unbiased genotyping allows the study of molecular mechanisms of lymphatic involvement of hemovascular disorders.


Assuntos
Mutação da Fase de Leitura , Anormalidades Linfáticas/genética , Anormalidades Linfáticas/patologia , Síndrome de Sturge-Weber/genética , Síndrome de Sturge-Weber/patologia , Proteína p120 Ativadora de GTPase/genética , Animais , Corantes/administração & dosagem , Modelos Animais de Doenças , Exoma/genética , Feminino , Humanos , Hiperplasia , Verde de Indocianina/administração & dosagem , Anormalidades Linfáticas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Síndrome de Sturge-Weber/metabolismo , Proteína p120 Ativadora de GTPase/metabolismo
6.
Am J Pathol ; 184(12): 3163-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25283357

RESUMO

Capillary malformation-arteriovenous malformation (CM-AVM) is an autosomal dominant blood vascular (BV) disorder characterized by CM and fast flow BV lesions. Inactivating mutations of the RASA1 gene are the cause of CM-AVM in most cases. RASA1 is a GTPase-activating protein that acts as a negative regulator of the Ras small GTP-binding protein. In addition, RASA1 performs Ras-independent functions in intracellular signal transduction. Whether CM-AVM results from loss of an ability of RASA1 to regulate Ras or loss of a Ras-independent function of RASA1 is unknown. To address this, we generated Rasa1 knockin mice with an R780Q point mutation that abrogates RASA1 catalytic activity specifically. Homozygous Rasa1(R780Q/R780Q) mice showed the same severe BV abnormalities as Rasa1-null mice and died midgestation. This finding indicates that BV abnormalities in CM-AVM develop as a result of loss of an ability of RASA1 to control Ras activation and not loss of a Ras-independent function of this molecule. More important, findings indicate that inhibition of Ras signaling is likely to represent an effective means of therapy for this disease.


Assuntos
Malformações Arteriovenosas/genética , Vasos Sanguíneos/anormalidades , Capilares/anormalidades , Mancha Vinho do Porto/genética , Proteína p120 Ativadora de GTPase/genética , Alelos , Animais , Catálise , Cruzamentos Genéticos , Análise Mutacional de DNA , Técnicas de Introdução de Genes , Homozigoto , Imuno-Histoquímica , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Mutação Puntual , Transdução de Sinais
7.
Blood ; 122(25): 4119-28, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24141370

RESUMO

Posttranscriptional modification of histones by methylation plays an important role in regulating Ag-driven T-cell responses. We have recently drawn correlations between allogeneic T-cell responses and the histone methyltransferase Ezh2, which catalyzes histone H3 lysine 27 trimethylation. The functional relevance of Ezh2 in T-cell alloimmunity remains unclear. Here, we identify a central role of Ezh2 in regulating allogeneic T-cell proliferation, differentiation, and function. Conditional loss of Ezh2 in donor T cells inhibited graft-versus-host disease (GVHD) in mice after allogeneic bone marrow (BM) transplantation. Although Ezh2-deficient T cells were initially activated to proliferate upon alloantigenic priming, their ability to undergo continual proliferation and expansion was defective during late stages of GVHD induction. This effect of Ezh2 ablation was largely independent of the proapoptotic molecule Bim. Unexpectedly, as a gene silencer, Ezh2 was required to promote the expression of transcription factors Tbx21 and Stat4. Loss of Ezh2 in T cells specifically impaired their differentiation into interferon (IFN)-γ-producing effector cells. However, Ezh2 ablation retained antileukemia activity in alloreactive T cells, leading to improved overall survival of the recipients. Our findings justify investigation of modulating Ezh2 as a therapeutic strategy for the treatment of GVHD and other T cell-mediated inflammatory disorders.


Assuntos
Epigênese Genética , Doença Enxerto-Hospedeiro/enzimologia , Complexo Repressor Polycomb 2/metabolismo , Linfócitos T/enzimologia , Aloenxertos , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Transplante de Medula Óssea , Proteína Potenciadora do Homólogo 2 de Zeste , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Complexo Repressor Polycomb 2/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Linfócitos T/patologia
8.
Stem Cells ; 32(3): 741-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24123360

RESUMO

SHP2 is a widely expressed protein tyrosine phosphatase required for signal transduction from multiple cell surface receptors. Gain and loss of function SHP2 mutations in humans are known to cause Noonan and LEOPARD syndromes, respectively, that are characterized by numerous pathological conditions including male infertility. Using conditional gene targeting in the mouse, we found that SHP2 is required for maintaining spermatogonial stem cells (SSCs) and the production of germ cells required for male fertility. After deleting SHP2, spermatogenesis was halted at the initial step during which transit-amplifying undifferentiated spermatogonia are produced from SSCs. In the absence of SHP2, proliferation of SSCs and undifferentiated spermatogonia was inhibited, thus germ cells cannot be replenished and SSCs cannot undergo renewal. However, germ cells beyond the undifferentiated spermatogonia stage of development at the time of SHP2 knockout were able to complete their maturation to become sperm. In cultures of SSCs and their progeny, inhibition of SHP2 activity reduced growth factor-mediated intracellular signaling that regulates SSC proliferation and cell fate. Inhibition of SHP2 also decreased the number of SSCs present in culture and caused SSCs to detach from supporting cells. Injection of mice with an SHP2 inhibitor blocked the production of germ cells from SSCs. Together, our studies show that SHP2 is essential for SSCs to maintain fertility and indicates that the pathogenesis of infertility in humans with SHP2 mutations is due to compromised SSC functions that block spermatogenesis.


Assuntos
Fertilidade , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Espermatogônias/citologia , Espermatogônias/enzimologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Envelhecimento , Animais , Adesão Celular , Contagem de Células , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Deleção de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Knockout , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Transdução de Sinais , Espermatogênese
9.
Proc Natl Acad Sci U S A ; 109(40): 16264-9, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22988097

RESUMO

MHC class II-expressing thymocytes and thymic epithelial cells can mediate CD4 T-cell selection resulting in functionally distinct thymocyte-selected CD4 (T-CD4) and epithelial-selected CD4 (E-CD4) T cells, respectively. However, little is known about how T-cell receptor (TCR) signaling influences the development of these two CD4 T-cell subsets. To study TCR signaling for T-CD4 T-cell development, we used a GFP reporter system of Nur77 in which GFP intensity directly correlates with TCR signaling strength. T-CD4 T cells expressed higher levels of GFP than E-CD4 T cells, suggesting that T-CD4 T cells received stronger TCR signaling than E-CD4 T cells during selection. Elimination of Ras GTPase-activating protein enhanced E-CD4 but decreased T-CD4 T-cell selection efficiency, suggesting a shift to negative selection. Conversely, the absence of IL-2-inducible T-cell kinase that causes poor E-CD4 T-cell selection due to insufficient TCR signaling improved T-CD4 T-cell generation, consistent with rescue from negative selection. Strong TCR signaling during T-CD4 T-cell development correlates with the expression of the transcription factor promyelocytic leukemia zinc finger protein. However, although modulation of the signaling strength affected the efficiency of T-CD4 T-cell development during positive and negative selection, the signaling strength is not as important for the effector function of T-CD4 T cells. These findings indicate that innate T-CD4 T cells, together with invariant natural killer T cells and γδ T cells, receive strong TCR signals during their development and that signaling requirements for the development and the effector functions are distinct.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Fatores de Transcrição Kruppel-Like/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Transplante de Medula Óssea , Epitélio/imunologia , Citometria de Fluxo , Proteínas de Fluorescência Verde , Fatores de Troca do Nucleotídeo Guanina/genética , Camundongos , Camundongos Knockout , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteínas Tirosina Quinases/genética , Especificidade do Receptor de Antígeno de Linfócitos T , Timócitos/citologia , Timócitos/imunologia
10.
J Immunol ; 187(1): 151-63, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21646295

RESUMO

Activation of the Ras small GTP-binding protein is necessary for normal T cell development and function. However, it is unknown which Ras GTPase-activating proteins (RasGAPs) inactivate Ras in T cells. We used a T cell-specific RASA1-deficient mouse model to investigate the role of the p120 RasGAP (RASA1) in T cells. Death of CD4(+)CD8(+) double-positive thymocytes was increased in RASA1-deficient mice. Despite this finding, on an MHC class II-restricted TCR transgenic background, evidence was obtained for increased positive selection of thymocytes associated with augmented activation of the Ras-MAPK pathway. In the periphery, RASA1 was found to be dispensable as a regulator of Ras-MAPK activation and T cell functional responses induced by full agonist peptides. However, numbers of naive T cells were substantially reduced in RASA1-deficient mice. Loss of naive T cells in the absence of RASA1 could be attributed in part to impaired responsiveness to the IL-7 prosurvival cytokine. These findings reveal an important role for RASA1 as a regulator of double-positive survival and positive selection in the thymus as well as naive T cell survival in the periphery.


Assuntos
Fase de Repouso do Ciclo Celular/imunologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Proteína p120 Ativadora de GTPase/fisiologia , Proteínas Ativadoras de ras GTPase/fisiologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fase de Repouso do Ciclo Celular/genética , Subpopulações de Linfócitos T/metabolismo , Timo/metabolismo , Proteína p120 Ativadora de GTPase/deficiência , Proteína p120 Ativadora de GTPase/genética , Proteínas Ativadoras de ras GTPase/deficiência , Proteínas Ativadoras de ras GTPase/genética
11.
Immunol Rev ; 232(1): 240-54, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19909368

RESUMO

Adapter proteins play key roles in intracellular signal transduction through complex formation with catalytically active signaling molecules. In T lymphocytes, the role of several different types of adapter proteins in T-cell antigen receptor signal transduction is well established. An exception to this is the family of T-cell-specific adapter (TSAd) proteins comprising of TSAd, adapter protein of unknown function (ALX), SH2D4A, and SH2D4B. Only recently has the function of these adapters in T-cell signal transduction been explored. Here, we discuss advances in our understanding of the role of this family of adapter proteins in T cells. Their function as regulators of signal transduction in other cell types is also discussed.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Membrana/metabolismo , Multimerização Proteica/imunologia , Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/imunologia , Camundongos , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/citologia
12.
J Exp Med ; 203(2): 281-7, 2006 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-16446380

RESUMO

T cell-specific adapter protein (TSAd) is a SRC-homology-2 (SH2) domain-containing intracellular signaling molecule that is required for T cell antigen receptor (TCR)-induced cytokine synthesis in T cells. How TSAd functions in TCR signal transduction is not clear. Previous work has suggested a nuclear role for this adapter. However, other evidence suggests that TSAd also functions in the cytoplasm. Using T cells from TSAd-deficient mice, we now show that the major role of TSAd in the cytoplasm is in activation of the LCK protein tyrosine kinase at the outset of TCR signal transduction. Consequently, TSAd regulates several downstream signaling events, including intracellular calcium mobilization and activation of the Ras-extracellular signal-regulated kinase signaling pathway. TSAd regulates LCK activity directly through physical interaction with LCK SH3 and SH2 domains. These studies reveal TSAd as a positive regulator of proximal TCR signal transduction and provide important new information on the mechanism of TCR-induced LCK activation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Subpopulações de Linfócitos T/enzimologia , Subpopulações de Linfócitos T/imunologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células Cultivadas , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia
13.
Methods Mol Biol ; 2319: 69-75, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34331244

RESUMO

There is increasing interest in the study of the mammalian lymphatic system, including the lymphatic endothelial cells (LECs) that make up lymphatic vessels. The ability to isolate primary LECs from tissue of normal and genetically modified mice permits detailed analysis of this unique cell type. Here, we describe a robust protocol for the isolation and in vitro expansion of LECs from mouse lung by antibody-based magnetic separation.


Assuntos
Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Células Endoteliais/citologia , Separação Imunomagnética/métodos , Pulmão/citologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Meios de Cultura/química , Células Endoteliais/metabolismo , Técnicas In Vitro , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos
14.
J Immunol ; 181(3): 2019-27, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18641339

RESUMO

T cell-specific adapter (TSAd) protein and adapter protein in lymphocytes of unknown function (ALX) are two related Src homology 2 (SH2) domain-containing signaling adapter molecules that have both been shown to regulate TCR signal transduction in T cells. TSAd is required for normal TCR-induced synthesis of IL-2 and other cytokines in T cells and acts at least in part by promoting activation of the LCK protein tyrosine kinase at the outset of the TCR signaling cascade. By contrast, ALX functions as a negative-regulator of TCR-induced IL-2 synthesis through as yet undetermined mechanisms. In this study, we report a novel T cell-expressed adapter protein named SH2D4A that contains an SH2 domain that is highly homologous to the TSAd protein and ALX SH2 domains and that shares other structural features with these adapters. To examine the function of SH2D4A in T cells we produced SH2D4A-deficient mice by homologous recombination in embryonic stem cells. T cell development, homeostasis, proliferation, and function were all found to be normal in these mice. Furthermore, knockdown of SH2D4A expression in human T cells did not impact upon their function. We conclude that in contrast to TSAd and ALX proteins, SH2D4A is dispensable for TCR signal transduction in T cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Listeriose/genética , Listeriose/imunologia , Listeriose/metabolismo , Proteínas de Membrana/química , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , RNA Interferente Pequeno/genética , Alinhamento de Sequência
15.
Nat Commun ; 11(1): 180, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31924779

RESUMO

Macropinocytosis is an evolutionarily-conserved, large-scale, fluid-phase form of endocytosis that has been ascribed different functions including antigen presentation in macrophages and dendritic cells, regulation of receptor density in neurons, and regulation of tumor growth under nutrient-limiting conditions. However, whether macropinocytosis regulates the expansion of non-transformed mammalian cells is unknown. Here we show that primary mouse and human T cells engage in macropinocytosis that increases in magnitude upon T cell activation to support T cell growth even under amino acid (AA) replete conditions. Mechanistically, macropinocytosis in T cells provides access of extracellular AA to an endolysosomal compartment to sustain activation of the mechanistic target of rapamycin complex 1 (mTORC1) that promotes T cell growth. Our results thus implicate a function of macropinocytosis in mammalian cell growth beyond Ras-transformed tumor cells via sustained mTORC1 activation.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Pinocitose/fisiologia , Linfócitos T/fisiologia , Aminoácidos , Animais , Linfócitos T CD4-Positivos/fisiologia , Endossomos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Linfócitos T/citologia
16.
J Clin Invest ; 129(9): 3545-3561, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31185000

RESUMO

Combined germline and somatic second hit inactivating mutations of the RASA1 gene, which encodes a negative regulator of the Ras signaling pathway, cause blood and lymphatic vascular lesions in the human autosomal dominant vascular disorder capillary malformation-arteriovenous malformation (CM-AVM). How RASA1 mutations in endothelial cells (EC) result in vascular lesions in CM-AVM is unknown. Here, using different murine models of RASA1-deficiency, we found that RASA1 was essential for the survival of EC during developmental angiogenesis in which primitive vascular plexuses are remodeled into hierarchical vascular networks. RASA1 was required for EC survival during developmental angiogenesis because it was necessary for export of collagen IV from EC and deposition in vascular basement membranes. In the absence of RASA1, dysregulated Ras mitogen-activated protein kinase (MAPK) signal transduction in EC resulted in impaired folding of collagen IV and its retention in the endoplasmic reticulum (ER) leading to EC death. Remarkably, the chemical chaperone, 4-phenylbutyric acid, and small molecule inhibitors of MAPK and 2-oxoglutarate dependent collagen IV modifying enzymes rescued ER retention of collagen IV and EC apoptosis and resulted in normal developmental angiogenesis. These findings have important implications with regards an understanding of the molecular pathogenesis of CM-AVM and possible means of treatment.


Assuntos
Colágeno Tipo IV/metabolismo , Vasos Linfáticos/embriologia , Proteína p120 Ativadora de GTPase/genética , Proteína p120 Ativadora de GTPase/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Malformações Arteriovenosas/metabolismo , Linhagem Celular Tumoral , Edema/metabolismo , Retículo Endoplasmático/metabolismo , Feminino , Valvas Cardíacas , Ventrículos do Coração/patologia , Hemorragia/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neovascularização Patológica , Fenótipo , Fenilbutiratos/farmacologia , Gravidez , Transdução de Sinais , Transgenes
17.
Eur J Med Genet ; 61(1): 11-16, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29024832

RESUMO

Capillary malformation-arteriovenous malformation (CM-AVM) is an autosomal dominant vascular disorder that is associated with inherited inactivating mutations of the RASA1 gene in the majority of cases. Characteristically, patients exhibit one or more focal cutaneous CM that may occur alone or together with AVM, arteriovenous fistulas or lymphatic vessel abnormalities. The focal nature and varying presentation of lesions has led to the hypothesis that somatic "second hit" inactivating mutations of RASA1 are necessary for disease development. In this study, we examined CM from four different CM-AVM patients for the presence of somatically acquired RASA1 mutations. All four patients were shown to possess inactivating heterozygous germline RASA1 mutations. In one of the patients, a somatic inactivating RASA1 mutation (c.1534C > T, p.Arg512*) was additionally identified in CM lesion tissue. The somatic RASA1 mutation was detected within endothelial cells specifically and was in trans with the germline RASA1 mutation. Together with the germline RASA1 mutation (c.2125C > T, p.Arg709*) in the same patient, the endothelial cell somatic RASA1 mutation likely contributed to lesion development. These studies provide the first clear evidence of the second hit model of CM-AVM pathogenesis.


Assuntos
Malformações Arteriovenosas/genética , Capilares/anormalidades , Células Endoteliais/metabolismo , Mancha Vinho do Porto/genética , Proteína p120 Ativadora de GTPase/genética , Adolescente , Adulto , Malformações Arteriovenosas/patologia , Capilares/patologia , Criança , Endotélio Vascular/metabolismo , Feminino , Mutação em Linhagem Germinativa , Humanos , Masculino , Mancha Vinho do Porto/patologia , Proteína p120 Ativadora de GTPase/metabolismo
18.
Genesis ; 45(12): 762-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18064675

RESUMO

p120 Ras GTPase-activating protein (RasGAP) encoded by the rasa1 gene in mice is a prototypical member of the RasGAP family of proteins involved in negative-regulation of the p21 Ras proto-oncogene. RasGAP has been implicated in signal transduction through a number of cell surface receptors. In humans, inactivating mutations in the coding region of the RASA1 gene cause capillary malformation arteriovenous malformation. In mice, generalized disruption of the rasa1 gene results in early embryonic lethality associated with defective vasculogenesis and increased apoptosis of neuronal cells. The early lethality in this mouse model precludes its use to further study the importance of RasGAP as a regulator of cell function. Therefore, to circumvent this problem, we have generated a conditional rasa1 knockout mouse. In this mouse, an exon that encodes a part of the RasGAP protein essential for catalytic activity has been flanked by loxP recognition sites. With the use of different constitutive and inducible Cre transgenic mouse lines, we show that deletion of this exon from the rasa1 locus results in effective loss of expression of catalytically-active RasGAP from a variety of adult tissues. The conditional rasa1 mouse will be useful for the analysis of the role of RasGAP in mature cell types.


Assuntos
Alelos , Genes Letais , Camundongos Knockout/genética , Proteína p120 Ativadora de GTPase/genética , Animais , Éxons , Camundongos , Mutação , Proto-Oncogene Mas , Deleção de Sequência
19.
J Clin Invest ; 127(7): 2569-2585, 2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28530642

RESUMO

Capillary malformation-arteriovenous malformation (CM-AVM) is a blood and lymphatic vessel (LV) disorder that is caused by inherited inactivating mutations of the RASA1 gene, which encodes p120 RasGAP (RASA1), a negative regulator of the Ras small GTP-binding protein. How RASA1 mutations lead to the LV leakage defects that occur in CM-AVM is not understood. Here, we report that disruption of the Rasa1 gene in adult mice resulted in loss of LV endothelial cells (LECs) specifically from the leaflets of intraluminal valves in collecting LVs. As a result, valves were unable to prevent fluid backflow and the vessels were ineffective pumps. Furthermore, disruption of Rasa1 in midgestation resulted in LEC apoptosis in developing LV valves and consequently failed LV valvulogenesis. Similar phenotypes were observed in induced RASA1-deficient adult mice and embryos expressing a catalytically inactive RASA1R780Q mutation. Thus, RASA1 catalytic activity is essential for the function and development of LV valves. These data provide a partial explanation for LV leakage defects and potentially other LV abnormalities observed in CM-AVM.


Assuntos
Apoptose , Células Endoteliais/metabolismo , Vasos Linfáticos/metabolismo , Mutação de Sentido Incorreto , Proteína p120 Ativadora de GTPase/metabolismo , Animais , Camundongos , Camundongos Mutantes , Proteína p120 Ativadora de GTPase/genética
20.
Clin Cancer Res ; 11(10): 3614-23, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15897556

RESUMO

Transporter associated with antigen processing (TAP), a member of the ATP-binding cassette transporter superfamily, is composed of two integral membrane proteins, TAP-1 and TAP-2. Each subunit has a C-terminal nucleotide-binding domain that binds and hydrolyzes ATP to energize peptide translocation across the endoplasmic reticulum membrane. A motif comprising the sequence LSGGQ (called the signature motif) and the amino acid that is immediately C-terminal to this motif are highly conserved in the nucleotide-binding domains of ATP-binding cassette transporters. To search for natural variants of TAP-1 with alterations in or near the signature motif, we sequenced the TAP-1 exon 10 amplified from 103 human colon cancer samples. We found a rare TAP-1 allele with an R>Q alteration at a residue immediately C-terminal to the signature motif (R648) that occurred 17.5 times more frequently in colon cancers with down-regulated surface class I MHC than those with normal MHC levels (P = 0.01). Functional analysis revealed that the Q648 variant had significantly reduced peptide translocation activity compared with TAP-1 (R648). In addition, we found that mutations S644R, G645R, G646S, and G646D interfered with TAP-1 activity. TAP-1 G646D, which showed the most severe defect, resided normally in the endoplasmic reticulum and associated with the peptide loading complex, but failed to transport peptide across the endoplasmic reticulum membrane. Thus, a TAP-1 polymorphism adjacent to the signature motif may be a contributing factor for MHC class I down-regulation in colon cancer. Given the widespread defects in DNA mismatch repair in colon cancer, mutations at or near the signature domain can potentially modulate antigen processing.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Neoplasias do Colo/genética , Antígenos de Histocompatibilidade Classe I/genética , Polimorfismo Genético , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Alelos , Sequência de Aminoácidos , Pareamento Incorreto de Bases , Neoplasias do Colo/patologia , Reparo do DNA , Regulação para Baixo , Genes MHC Classe I , Antígenos HLA/biossíntese , Humanos , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA