Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
JCI Insight ; 7(4)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35015735

RESUMO

Capillary malformation-arteriovenous malformation (CM-AVM) is a blood vascular anomaly caused by inherited loss-of-function mutations in RASA1 or EPHB4 genes, which encode p120 Ras GTPase-activating protein (p120 RasGAP/RASA1) and Ephrin receptor B4 (EPHB4). However, whether RASA1 and EPHB4 function in the same molecular signaling pathway to regulate the blood vasculature is uncertain. Here, we show that induced endothelial cell-specific (EC-specific) disruption of Ephb4 in mice resulted in accumulation of collagen IV in the EC ER, leading to EC apoptotic death and defective developmental, neonatal, and pathological angiogenesis, as reported previously in induced EC-specific RASA1-deficient mice. Moreover, defects in angiogenic responses in EPHB4-deficient mice could be rescued by drugs that inhibit signaling through the Ras pathway and drugs that promote collagen IV export from the ER. However, EPHB4-mutant mice that expressed a form of EPHB4 that is unable to physically engage RASA1 but retains protein tyrosine kinase activity showed normal angiogenic responses. These findings provide strong evidence that RASA1 and EPHB4 function in the same signaling pathway to protect against the development of CM-AVM independent of physical interaction and have important implications for possible means of treatment of this disease.


Assuntos
Colágeno Tipo IV/metabolismo , DNA/genética , Células Endoteliais/patologia , Mutação , Neovascularização Patológica/genética , Receptor EphB4/genética , Malformações Vasculares/genética , Animais , Células Cultivadas , Análise Mutacional de DNA , Células Endoteliais/metabolismo , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Receptor EphB4/metabolismo , Malformações Vasculares/metabolismo , Malformações Vasculares/patologia , Proteína p120 Ativadora de GTPase/deficiência
2.
Cell Mol Gastroenterol Hepatol ; 12(4): 1479-1502, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34242819

RESUMO

BACKGROUND & AIMS: CD4+ T cells are regulated by activating and inhibitory cues, and dysregulation of these proper regulatory inputs predisposes these cells to aberrant inflammation and exacerbation of disease. We investigated the role of the inhibitory receptor paired immunoglobulin-like receptor B (PIR-B) in the regulation of the CD4+ T-cell inflammatory response and exacerbation of the colitic phenotype. METHODS: We used Il10-/- spontaneous and CD4+CD45RBhi T-cell transfer models of colitis with PIR-B-deficient (Pirb-/-) mice. Flow cytometry, Western blot, and RNA sequencing analysis was performed on wild-type and Pirb-/- CD4+ T cells. In silico analyses were performed on RNA sequencing data set of ileal biopsy samples from pediatric CD and non-inflammatory bowel disease patients and sorted human memory CD4+ T cells. RESULTS: We identified PIR-B expression on memory CD4+ interleukin (IL)17a+ cells. We show that PIR-B regulates CD4+ T-helper 17 cell (Th17)-dependent chronic intestinal inflammatory responses and the development of colitis. Mechanistically, we show that the PIR-B- Src-homology region 2 domain-containing phosphatase-1/2 axis tempers mammalian target of rapamycin complex 1 signaling and mammalian target of rapamycin complex 1-dependent caspase-3/7 apoptosis, resulting in CD4+ IL17a+ cell survival. In silico analyses showed enrichment of transcriptional signatures for Th17 cells (RORC, RORA, and IL17A) and tissue resident memory (HOBIT, IL7R, and BLIMP1) networks in PIR-B+ murine CD4+ T cells and human CD4+ T cells that express the human homologue leukocyte immunoglobulin-like receptor subfamily B member 3 (LILRB3). High levels of LILRB3 expression were associated strongly with mucosal injury and a proinflammatory Th17 signature, and this signature was restricted to a treatment-naïve, severe pediatric CD population. CONCLUSIONS: Our findings show an intrinsic role for PIR-B/LILRB3 in the regulation of CD4+ IL17a+ T-cell pathogenic memory responses.


Assuntos
Regulação da Expressão Gênica , Imunomodulação , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Receptores Imunológicos/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Biomarcadores , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Colite/etiologia , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Imuno-Histoquímica , Memória Imunológica , Imunofenotipagem , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Receptores Imunológicos/metabolismo , Transdução de Sinais
3.
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
4.
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
5.
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
6.
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
7.
PLoS One ; 9(11): e112548, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25383712

RESUMO

The lymphatic vasculature plays a critical role in a number of disease conditions of increasing prevalence, such as autoimmune disorders, obesity, blood vascular diseases, and cancer metastases. Yet, unlike the blood vasculature, the tools available to interrogate the molecular basis of lymphatic dysfunction/disease have been lacking. More recently, investigators have reported that dysregulation of the PI3K pathway is involved in syndromic human diseases that involve abnormal lymphatic vasculatures, but there have been few compelling results that show the direct association of this molecular pathway with lymphatic dysfunction in humans. Using near-infrared fluorescence lymphatic imaging (NIRFLI) to phenotype and next generation sequencing (NGS) for unbiased genetic discovery in a family with non-syndromic lymphatic disease, we discovered a rare, novel mutation in INPPL1 that encodes the protein SHIP2, which is a negative regulator of the PI3K pathway, to be associated with lymphatic dysfunction in the family. In vitro interrogation shows that SHIP2 is directly associated with impairment of normal lymphatic endothelial cell (LEC) behavior and that SHIP2 associates with receptors that are associated in lymphedema, implicating its direct involvement in the lymphatic vasculature.


Assuntos
Fator de Crescimento de Hepatócito/genética , Linfedema/genética , Linfedema/patologia , MAP Quinase Quinase Quinases/genética , Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/genética , Domínios de Homologia de src , Adulto , Idoso , Células Cultivadas , Células Endoteliais/patologia , Feminino , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Pessoa de Meia-Idade , Imagem Óptica/métodos , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Mutação Puntual , Análise de Sequência de DNA
8.
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
9.
Bone ; 69: 55-60, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25178522

RESUMO

Src homology-2 protein tyrosine phosphatase (SHP-2) that is encoded by the PTPN11 gene in humans is an intracellular signaling molecule that couples growth factor receptors to activation of the Ras small GTP-binding protein that regulates cell growth, proliferation and differentiation. Germline mutations of PTPN11 are causative of Noonan syndrome and LEOPARD syndrome in humans in which there are recognized skeletal abnormalities that include growth retardation, spinal curvature and chest malformations. In addition, combined somatic and germline PTPN11 mutations have been shown to be responsible for a rare benign bone cartilaginous tumor disease known as metachondromatosis. In parallel, gene targeting studies performed in mice have revealed an essential role for SHP-2 as a regulator of bone and skeletal development. In this review the significance of these findings in mice to the understanding of the pathogenesis of skeletal abnormalities in humans with SHP-2 mutations is discussed.


Assuntos
Desenvolvimento Ósseo/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Animais , Humanos , Síndrome LEOPARD/genética , Camundongos , Síndrome de Noonan/genética
10.
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
11.
J Bone Miner Res ; 29(3): 761-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23929766

RESUMO

Metachondromatosis is a benign bone disease predominantly observed in the hands and feet of children or young adults demonstrating two different manifestations: a cartilage-capped bony outgrowth on the surface of the bone called exostosis and ectopic cartilaginous nodules inside the bone called enchondroma. Recently, it has been reported that loss-of-function mutations of the SHP2 gene, which encodes the SHP2 protein tyrosine phosphatase, are associated with metachondromatosis. The purpose of this study was to investigate the role of SHP2 in postnatal cartilage development, which is largely unknown. We disrupted Shp2 during the postnatal stage of mouse development in a chondrocyte-specific manner using a tamoxifen-inducible system. We found tumor-like nodules on the hands and feet within a month after the initial induction. The SHP2-deficient mice demonstrated an exostosis-like and enchondroma-like phenotype in multiple bones of the hands, feet, and ribs as assessed by X-ray and micro-computed tomography (CT). Histological assessment revealed the disorganization of the growth plate cartilage, a cartilaginous protrusion from the epiphyseal bone, and ectopic cartilage nodules within the bones, which is consistent with the pathological features of metachondromatosis in humans (ie, both exostosis and enchondroma). At molecular levels, we observed an abundant expression of Indian hedgehog protein (IHH) and fibroblast growth factor 2 (FGF2) and impaired expression of mitogen-activated protein kinases (MAPK) in the affected cartilage nodules in the SHP2-deficient mice. In summary, we have generated a mouse model of metachondromatosis that includes manifestations of exostosis and enchondroma. This study provides a novel model for the investigation of the pathophysiology of the disease and advances the understanding of metachondromatosis. This model will be useful to identify molecular mechanisms for the disease cause and progression as well as to develop new therapeutic strategies in the future.


Assuntos
Neoplasias Ósseas/patologia , Cartilagem/metabolismo , Condrócitos/citologia , Condromatose/patologia , Exostose Múltipla Hereditária/patologia , Domínios de Homologia de src/genética , Animais , Neoplasias Ósseas/metabolismo , Condrócitos/metabolismo , Condromatose/metabolismo , Exostose Múltipla Hereditária/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
12.
Dis Model Mech ; 6(6): 1448-58, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24077964

RESUMO

In mice, induced global disruption of the Ptpn11 gene, which encodes the SHP-2 tyrosine phosphatase, results in severe skeletal abnormalities. To understand the extent to which skeletal abnormalities can be attributed to perturbation of SHP-2 function in bone-forming osteoblasts and chondrocytes, we generated mice in which disruption of Ptpn11 is restricted to mesenchymal stem cells (MSCs) and their progeny, which include both cell types. MSC-lineage-specific SHP-2 knockout (MSC SHP-2 KO) mice exhibited postnatal growth retardation, limb and chest deformity, and calvarial defects. These skeletal abnormalities were associated with an absence of mature osteoblasts and massive chondrodysplasia with a vast increase in the number of terminally differentiated hypertrophic chondrocytes in affected bones. Activation of mitogen activated protein kinases (MAPKs) and protein kinase B (PKB; also known as AKT) was impaired in bone-forming cells of MSC SHP-2 KO mice, which provides an explanation for the skeletal defects that developed. These findings reveal a cell-autonomous role for SHP-2 in bone-forming cells in mice in the regulation of skeletal development. The results add to our understanding of the pathophysiology of skeletal abnormalities observed in humans with germline mutations in the PTPN11 gene (e.g. Noonan syndrome and LEOPARD syndrome).


Assuntos
Osso e Ossos/anormalidades , Retardo do Crescimento Fetal/genética , Deformidades Congênitas dos Membros/genética , Células-Tronco Mesenquimais/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Tórax/anormalidades , Animais , Camundongos , Camundongos Knockout , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética
13.
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
14.
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
15.
J Immunol ; 190(11): 5818-28, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23636056

RESUMO

Graft-versus-host disease (GVHD) induced by donor-derived T cells remains the major limitation of allogeneic bone marrow transplantation (allo-BMT). We previously reported that the pan-Notch inhibitor dominant-negative form of Mastermind-like 1 (DNMAML) markedly decreased the severity and mortality of acute GVHD mediated by CD4(+) T cells in mice. To elucidate the mechanisms of Notch action in GVHD and its role in CD8(+) T cells, we studied the effects of Notch inhibition in alloreactive CD4(+) and CD8(+) T cells using mouse models of allo-BMT. DNMAML blocked GVHD induced by either CD4(+) or CD8(+) T cells. Both CD4(+) and CD8(+) Notch-deprived T cells had preserved expansion in lymphoid organs of recipients, but profoundly decreased IFN-γ production despite normal T-bet and enhanced Eomesodermin expression. Alloreactive DNMAML T cells exhibited decreased Ras/MAPK and NF-κB activity upon ex vivo restimulation through the TCR. In addition, alloreactive T cells primed in the absence of Notch signaling had increased expression of several negative regulators of T cell activation, including Dgka, Cblb, and Pdcd1. DNMAML expression had modest effects on in vivo proliferation but preserved overall alloreactive T cell expansion while enhancing accumulation of pre-existing natural regulatory T cells. Overall, DNMAML T cells acquired a hyporesponsive phenotype that blocked cytokine production but maintained their expansion in irradiated allo-BMT recipients, as well as their in vivo and ex vivo cytotoxic potential. Our results reveal parallel roles for Notch signaling in alloreactive CD4(+) and CD8(+) T cells that differ from past reports of Notch action and highlight the therapeutic potential of Notch inhibition in GVHD.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Receptores Notch/antagonistas & inibidores , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Citotoxicidade Imunológica , Ativação Enzimática , Regulação da Expressão Gênica , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/metabolismo , Interferon gama/biossíntese , Ativação Linfocitária , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores Notch/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
16.
Mol Immunol ; 55(3-4): 292-302, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23522726

RESUMO

TCR-mediated activation of the Ras signaling pathway is critical for T cell development in the thymus and function in the periphery. However, which members of a family of Ras GTPase-activating proteins (RasGAPs) negatively regulate Ras activation in T cells is unknown. In this study we examined a potential function for the neurofibromin 1 (NF1) RasGAP in the T cell lineage with the use of T cell-specific NF1-deficient mice. Surprisingly, on an MHC class I-restricted TCR transgenic background, NF1 was found to promote thymocyte positive selection. By contrast, NF1 neither promoted nor inhibited the negative selection of thymocytes. In the periphery, NF1 was found to be necessary for the maintenance of normal numbers of naïve CD4⁺ and CD8⁺ T cells but was dispensable as a regulator of TCR-induced Ras activation, cytokine synthesis, proliferation and differentiation and death. These findings point to a novel unexpected role for NF1 in T cell development as well as a regulator of T cell homeostasis.


Assuntos
Diferenciação Celular/imunologia , Neurofibromina 1/fisiologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Proteínas ras/metabolismo , Animais , Seleção Clonal Mediada por Antígeno/imunologia , Feminino , Ativação Linfocitária/imunologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurofibromina 1/deficiência , Neurofibromina 1/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/citologia
17.
Sci Signal ; 6(264): re1, 2013 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-23443682

RESUMO

Inactivation of the small guanosine triphosphate-binding protein Ras during receptor signal transduction is mediated by Ras guanosine triphosphatase (GTPase)-activating proteins (RasGAPs). Ten different RasGAPs have been identified and have overlapping patterns of tissue distribution. However, genetic analyses are revealing critical nonredundant functions for each RasGAP in tissue homeostasis and as regulators of disease processes in mouse and man. Here, we discuss advances in understanding the role of RasGAPs in the maintenance of tissue integrity.


Assuntos
Homeostase/fisiologia , Modelos Biológicos , Transdução de Sinais/fisiologia , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/fisiologia , Proteínas ras/fisiologia , Animais , Membrana Celular/metabolismo , Humanos , Camundongos , Neurofibromina 1/fisiologia , Estrutura Terciária de Proteína , Proteína p120 Ativadora de GTPase/fisiologia
18.
Proc Natl Acad Sci U S A ; 110(1): E79-88, 2013 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-23236157

RESUMO

The molecular mechanism underlying adipogenesis and the physiological functions of adipose tissue are not fully understood. We describe here a unique mouse model of severe lipodystrophy. Ablation of Ptpn11/Shp2 in adipocytes, mediated by aP2-Cre, led to premature death, lack of white fat, low blood pressure, compensatory erythrocytosis, and hepatic steatosis in Shp2(fat-/-) mice. Fat transplantation partially rescued the lifespan and blood pressure in Shp2(fat-/-) mice, and administration of leptin also restored partially the blood pressure of mutant animals with endogenous leptin deficiency. Consistently, homozygous deletion of Shp2 inhibited adipocyte differentiation from embryonic stem (ES) cells. Biochemical analyses suggest a Shp2-TAO2-p38-p300-PPARγ pathway in adipogenesis, in which Shp2 suppresses p38 activation, leading to stabilization of p300 and enhanced PPARγ expression. Inhibition of p38 restored adipocyte differentiation from Shp2(-/-) ES cells, and p38 signaling is also suppressed in obese patients and obese animals. These results illustrate an essential role of adipose tissue in mammalian survival and physiology and also suggest a common signaling mechanism involved in adipogenesis and obesity development.


Assuntos
Adipogenia/fisiologia , Modelos Animais de Doenças , Lipodistrofia/fisiopatologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Tecido Adiposo/transplante , Animais , Pressão Sanguínea/efeitos dos fármacos , Primers do DNA/genética , Proteína p300 Associada a E1A/metabolismo , Deleção de Genes , Leptina/administração & dosagem , Leptina/deficiência , Leptina/farmacologia , Camundongos , Camundongos Knockout , PPAR gama/metabolismo , Proteínas Quinases/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
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
20.
J Clin Invest ; 122(2): 733-47, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22232212

RESUMO

RASA1 (also known as p120 RasGAP) is a Ras GTPase-activating protein that functions as a regulator of blood vessel growth in adult mice and humans. In humans, RASA1 mutations cause capillary malformation-arteriovenous malformation (CM-AVM); whether it also functions as a regulator of the lymphatic vasculature is unknown. We investigated this issue using mice in which Rasa1 could be inducibly deleted by administration of tamoxifen. Systemic loss of RASA1 resulted in a lymphatic vessel disorder characterized by extensive lymphatic vessel hyperplasia and leakage and early lethality caused by chylothorax (lymphatic fluid accumulation in the pleural cavity). Lymphatic vessel hyperplasia was a consequence of increased proliferation of lymphatic endothelial cells (LECs) and was also observed in mice in which induced deletion of Rasa1 was restricted to LECs. RASA1-deficient LECs showed evidence of constitutive activation of Ras in situ. Furthermore, in isolated RASA1-deficient LECs, activation of the Ras signaling pathway was prolonged and cellular proliferation was enhanced after ligand binding to different growth factor receptors, including VEGFR-3. Blockade of VEGFR-3 was sufficient to inhibit the development of lymphatic vessel hyperplasia after loss of RASA1 in vivo. These findings reveal a role for RASA1 as a physiological negative regulator of LEC growth that maintains the lymphatic vasculature in a quiescent functional state through its ability to inhibit Ras signal transduction initiated through LEC-expressed growth factor receptors such as VEGFR-3.


Assuntos
Células Endoteliais/fisiologia , Doenças Linfáticas/patologia , Vasos Linfáticos/patologia , Vasos Linfáticos/fisiologia , Proteína p120 Ativadora de GTPase/metabolismo , Animais , Proliferação de Células , Células Endoteliais/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Hiperplasia , Linfangiogênese/fisiologia , Doenças Linfáticas/fisiopatologia , Vasos Linfáticos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteína p120 Ativadora de GTPase/genética , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA