Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Eur J Immunol ; 52(2): 261-269, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731490

RESUMO

The differentiation of T cells from lymphoid progenitors in the thymus follows sequential developmental stages that constantly require interaction with thymic epithelial cells. Several distinct aspects of early T cell development depend on the activation of Notch receptors on thymocytes, while the selection of thymocytes at later stages are believed to be Notch independent. Using reverse genetic approaches and whole-thymus live imaging in an in vivo teleost model, the medaka, we report that Notch1 signals is required for proliferation and specification of developing T cells as well as involved in their selection in the thymus. We reveal that Notch1 controls the migratory behavior of thymocytes through controlling the chemokine receptor Ccr9b and thereby influence the T cell receptor (TCR) activation. Hence, we propose that, in lower vertebrates, the function of Notch signaling extends to all stages of T cell development, except when thymocytes undergo TCRß rearrangement.


Assuntos
Movimento Celular , Proteínas de Peixes/imunologia , Oryzias , Receptor Notch1/deficiência , Transdução de Sinais , Linfócitos T/imunologia , Timo/imunologia , Animais , Movimento Celular/genética , Movimento Celular/imunologia , Proteínas de Peixes/deficiência , Oryzias/genética , Oryzias/imunologia , Receptor Notch1/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
2.
Development ; 147(12)2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32467240

RESUMO

The cortical and medullary thymic epithelial cell (cTEC and mTEC) lineages are essential for inducing T cell lineage commitment, T cell positive selection and the establishment of self-tolerance, but the mechanisms controlling their fetal specification and differentiation are poorly understood. Here, we show that notch signaling is required to specify and expand the mTEC lineage. Notch1 is expressed by and active in TEC progenitors. Deletion of Notch1 in TECs resulted in depletion of mTEC progenitors and dramatic reductions in mTECs during fetal stages, consistent with defects in mTEC specification and progenitor expansion. Conversely, forced notch signaling in all TECs resulted in widespread expression of mTEC progenitor markers and profound defects in TEC differentiation. In addition, lineage-tracing analysis indicated that all mTECs have a history of receiving a notch signal, consistent with notch signaling occurring in mTEC progenitors. These data provide strong evidence for a requirement for notch signaling in specification of the mTEC lineage.


Assuntos
Desenvolvimento Fetal/genética , Receptor Notch1/metabolismo , Timo/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese , Receptor Notch1/deficiência , Receptor Notch1/genética , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo , Timo/citologia , Timo/crescimento & desenvolvimento
3.
Med Sci Monit ; 26: e919566, 2020 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-31964857

RESUMO

BACKGROUND Chemotherapy is widely used in gastric cancer treatment, but multidrug resistance remains a leading cause of chemotherapy failure. Trop2 is highly expressed in gastric tumor tissues and greatly influences cancer progression. However, little is known about the relationship between Trop2 and drug resistance in gastric cancer. MATERIAL AND METHODS In the present study, Trop2 was knocked down in BGC823 cells and overexpressed in HGC27. CCK-8 assay was performed to explore the relationship of Trop2 expression and cell proliferation treated with anticancer drugs. Flow cytometry was performed to assess the relationship between Trop2 and cell apoptosis after chemotherapy. Subcutaneous xenograft models were generated to explore the curative effect of DDP to GC in vivo. MRP1 and Notch1 expressions were assessed by Western blot. RESULTS Trop2 decreased cell proliferation inhibition and apoptosis after chemotherapeutic treatments. DDP showed stronger therapeutic effects on Trop2-knockdown tumor than control in vivo. MRP1 and Notch1 signaling pathway were confirmed to participate in Trop2-induced drug resistance. CONCLUSIONS Our findings suggest that Trop2 promotes the resistance of gastric cancer to chemotherapy by activating the Notch1 pathway.


Assuntos
Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , RNA Longo não Codificante/metabolismo , RNA Interferente Pequeno/farmacologia , Receptor Notch1/deficiência , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/patologia
4.
Cardiovasc Res ; 116(8): 1473-1486, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31591643

RESUMO

AIMS: Sinus venous valve (SVV) and sinoatrial node (SAN) develop together at the sinoatrial junction during embryogenesis. SVV ensures unidirectional cardiac input and SAN generates sinus rhythmic contraction, respectively; both functions are essential for embryonic survival. We aim to reveal the potential role of endocardial NOTCH signalling in SVV and SAN formation. METHODS AND RESULTS: We specifically deleted Notch1 in the endocardium using an Nfatc1Cre line. This deletion resulted in underdeveloped SVV and SAN, associated with reduced expression of T-box transcription factors, Tbx5 andTbx18, which are essential for the formation of SVV and SAN. The deletion also led to decreased expression of Wnt2 in myocardium of SVV and SAN. WNT2 treatment was able to rescue the growth defect of SVV and SAN resulted from the Notch1 deletion in whole embryo cultures. Furthermore, the Notch1 deletion reduced the expression of Nrg1 in the SVV myocardium and supplement of NRG1 restored the growth of SVV in cultured Notch1 knockout embryos. CONCLUSION: Our findings support that endocardial NOTCH1 controls the development of SVV and SAN by coordinating myocardial WNT and NRG1 signalling functions.


Assuntos
Seio Coronário/metabolismo , Miocárdio/metabolismo , Receptor Notch1/metabolismo , Nó Sinoatrial/metabolismo , Válvulas Venosas/metabolismo , Animais , Seio Coronário/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Knockout , Morfogênese , Neuregulina-1/genética , Neuregulina-1/metabolismo , Receptor Notch1/deficiência , Receptor Notch1/genética , Nó Sinoatrial/embriologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Válvulas Venosas/embriologia , Via de Sinalização Wnt , Proteína Wnt2/genética , Proteína Wnt2/metabolismo
5.
Stem Cells ; 37(7): 865-875, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30941836

RESUMO

Cancer stem cells (CSCs) play critical roles in cancer initiation, metastasis, recurrence, and drug resistance. Recent studies have revealed involvement of cancer-associated fibroblasts (CAFs) in regulating CSCs. However, the intracellular molecular mechanisms that determine the regulatory role of CAFs in modulating the plasticity of CSCs remain unknown. Here, we uncovered that intracellular Notch1 signaling in CAFs serves as a molecular switch, which modulates tumor heterogeneity and aggressiveness by inversely controlling stromal regulation of the plasticity and stemness of CSCs. Using mesenchymal stem cell-derived fibroblasts (MSC-DF) harboring reciprocal loss-of-function and gain-of-function Notch1 signaling, we found that MSC-DFNotch1-/- prompted cocultured melanoma cells to form more spheroids and acquire the phenotype (CD271+ and Nestin+ ) of melanoma stem/initiating cells (MICs), whereas MSC-DFN1IC+/+ suppressed melanoma cell sphere formation and mitigated properties of MICs. MSC-DFNotch1-/- increased stemness of CD271+ MIC, which resultantly exhibited stronger aggressiveness in vitro and in vivo, by upregulating Sox2/Oct4/Nanog expression. Consistently, when cografted with melanoma cells into NOD scid gamma (NSG) mice, MSC-DFNotch1-/- increased, but MSC-DFN1IC+/+ decreased, the amounts of CD271+ MIC in melanoma tissue. The amounts of CD271+ MIC regulated by MSC-DF carrying high or low Notch1 pathway activity is well correlated with capability of melanoma metastasis, supporting that melanoma metastasis is MIC-mediated. Our data demonstrate that intracellular Notch1 signaling in CAFs is a molecular switch dictating the plasticity and stemness of MICs, thereby regulating melanoma aggressiveness, and therefore that targeting the intracellular Notch1 signaling pathway in CAFs may present a new therapeutic strategy for melanoma. Stem Cells 2019;37:865-875.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptor Notch1/genética , Neoplasias Cutâneas/genética , Adapaleno/metabolismo , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Fibroblastos Associados a Câncer/patologia , Movimento Celular , Proliferação de Células , Técnicas de Cocultura , Xenoenxertos , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos SCID , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Células-Tronco Neoplásicas/patologia , Nestina/genética , Nestina/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Receptor Notch1/deficiência , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
6.
Circ Res ; 124(2): 211-224, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30582451

RESUMO

RATIONALE: Maintaining endothelial cells (EC) as a monolayer in the vessel wall depends on their metabolic state and gene expression profile, features influenced by contact with neighboring cells such as pericytes and smooth muscle cells (SMC). Failure to regenerate a normal EC monolayer in response to injury can result in occlusive neointima formation in diseases such as atherosclerosis and pulmonary arterial hypertension. OBJECTIVE: We investigated the nature and functional importance of contact-dependent communication between SMC and EC to maintain EC integrity. METHODS AND RESULTS: We found that in SMC and EC contact cocultures, BMPR2 (bone morphogenetic protein receptor 2) is required by both cell types to produce collagen IV to activate ILK (integrin-linked kinase). This enzyme directs p-JNK (phospho-c-Jun N-terminal kinase) to the EC membrane, where it stabilizes presenilin1 and releases N1ICD (Notch1 intracellular domain) to promote EC proliferation. This response is necessary for EC regeneration after carotid artery injury. It is deficient in EC-SMC Bmpr2 double heterozygous mice in association with reduced collagen IV production, decreased N1ICD, and attenuated EC proliferation, but can be rescued by targeting N1ICD to EC. Deletion of EC- Notch1 in transgenic mice worsens hypoxia-induced pulmonary hypertension, in association with impaired EC regenerative function associated with loss of precapillary arteries. We further determined that N1ICD maintains EC proliferative capacity by increasing mitochondrial mass and by inducing the phosphofructokinase PFKFB3 (fructose-2,6-bisphosphatase 3). Chromatin immunoprecipitation sequencing analyses showed that PFKFB3 is required for citrate-dependent H3K27 acetylation at enhancer sites of genes regulated by the acetyl transferase p300 and by N1ICD or the N1ICD target MYC and necessary for EC proliferation and homeostasis. CONCLUSIONS: Thus, SMC-EC contact is required for activation of Notch1 by BMPR2, to coordinate metabolism with chromatin remodeling of genes that enable EC regeneration, and to maintain monolayer integrity and vascular homeostasis in response to injury.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Lesões das Artérias Carótidas/metabolismo , Comunicação Celular , Proliferação de Células , Células Endoteliais/metabolismo , Metabolismo Energético , Epigênese Genética , Hipertensão Pulmonar/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor Notch1/metabolismo , Adulto , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Células Cultivadas , Montagem e Desmontagem da Cromatina , Técnicas de Cocultura , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Masculino , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Receptor Notch1/deficiência , Receptor Notch1/genética , Transdução de Sinais , Remodelação Vascular , Adulto Jovem
7.
Prog Neuropsychopharmacol Biol Psychiatry ; 84(Pt A): 63-70, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29410010

RESUMO

Neural oscillations and their interactions are associated with the coordination of neural groups, which provide a mechanism underlying information processing of brain functions. Notch1 receptor is involved in the neurological and psychiatric disorders, such as neurodevelopmental deficits, cerebral ischemia, Alzheimer's disease and depression. Here, we investigated the dynamics of neural oscillations in hippocampus of Notch1+/- mice in urethane-anesthetized state. Notch1 knockdown altered the distribution of power in the hippocampal DG areas, reduced theta (3-8 Hz) power and enhanced low gamma (LG, 30-50 Hz) and high gamma (HG, 50-100 Hz) power. Moreover, theta-gamma phase-amplitude coupling in the hippocampal DG area was markedly impaired in the Notch1+/- mice. The data further showed that the expression of NR2B was decreased, and the expressions of GABAARα1, GAD67 and parvalbumin were considerably increased after Notch1 knockdown. Taken together, our results suggest that Notch1 genetic deficiency significantly impaired the corss-frequency coupling of neural oscillations, and their interactions in the hippocampal DG region by means of disrupting the balance of excitatory and inhibitory receptors, which could be an underlying mechanism of cognitive impairment in neuropsychiatric disorders.


Assuntos
Giro Denteado/metabolismo , Ritmo Gama/fisiologia , Receptor Notch1/deficiência , Ritmo Teta/fisiologia , Anestésicos Intravenosos/farmacologia , Animais , Sincronização Cortical/fisiologia , Expressão Gênica/fisiologia , Glutamato Descarboxilase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor Notch1/genética , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Uretana/farmacologia
8.
PLoS One ; 13(12): e0203100, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30596653

RESUMO

During vertebrate cardiac development NOTCH signaling activity in the endocardium is essential for the crosstalk between endocardium and myocardium that initiates ventricular trabeculation and valve primordium formation. This crosstalk leads later to the maturation and compaction of the ventricular chambers and the morphogenesis of the cardiac valves, and its alteration may lead to disease. Although endocardial NOTCH signaling has been shown to be crucial for heart development, its physiological role in the myocardium has not been clearly established. Here we have used mouse genetics to evaluate the role of NOTCH in myocardial development. We have inactivated the unique and ubiquitous NOTCH effector RBPJ in early cardiomyocytes progenitors, and examined its consequences in cardiac development and function. Our results show that mice with Tnnt2-Cre-mediated myocardial-specific deletion of Rbpj develop to term, with homozygous mutant animals showing normal expression of cardiac development markers, and normal adult heart function. Similar observations have been obtained after Notch1 deletion with Tnnt2-Cre. We have also deleted Rbpj in both myocardial and endocardial progenitor cells, using the Nkx2.5-Cre driver, resulting in ventricular septal defect (VSD), double outlet right ventricle (DORV), and bicuspid aortic valve (BAV), due to NOTCH signaling abrogation in the endocardium of cardiac valves. Our data demonstrate that NOTCH-RBPJ inactivation in the myocardium does not affect heart development or adult cardiac function.


Assuntos
Deleção de Genes , Cardiopatias Congênitas , Coração/embriologia , Miocárdio/metabolismo , Receptor Notch1/deficiência , Transdução de Sinais , Proteínas rab de Ligação ao GTP/deficiência , Animais , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Camundongos , Camundongos Knockout , Miocárdio/patologia
9.
Development ; 144(14): 2629-2639, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28619820

RESUMO

Arterial specification and differentiation are influenced by a number of regulatory pathways. While it is known that the Vegfa-Notch cascade plays a central role, the transcriptional hierarchy controlling arterial specification has not been fully delineated. To elucidate the direct transcriptional regulators of Notch receptor expression in arterial endothelial cells, we used histone signatures, DNaseI hypersensitivity and ChIP-seq data to identify enhancers for the human NOTCH1 and zebrafish notch1b genes. These enhancers were able to direct arterial endothelial cell-restricted expression in transgenic models. Genetic disruption of SoxF binding sites established a clear requirement for members of this group of transcription factors (SOX7, SOX17 and SOX18) to drive the activity of these enhancers in vivo Endogenous deletion of the notch1b enhancer led to a significant loss of arterial connections to the dorsal aorta in Notch pathway-deficient zebrafish. Loss of SoxF function revealed that these factors are necessary for NOTCH1 and notch1b enhancer activity and for correct endogenous transcription of these genes. These findings position SoxF transcription factors directly upstream of Notch receptor expression during the acquisition of arterial identity in vertebrates.


Assuntos
Artérias/embriologia , Artérias/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Malformações Arteriovenosas/embriologia , Malformações Arteriovenosas/genética , Malformações Arteriovenosas/metabolismo , Elementos Facilitadores Genéticos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Gravidez , Receptor Notch1/deficiência , Fatores de Transcrição SOXF/deficiência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
FASEB J ; 31(10): 4347-4358, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28611114

RESUMO

It is well known that Notch1 signaling plays a crucial role in embryonic neural development and adult neurogenesis. The latest evidence shows that Notch1 also plays a critical role in synaptic plasticity in mature hippocampal neurons. So far, deeper insights into the function of Notch1 signaling during the different steps of adult neurogenesis are still lacking, and the mechanisms by which Notch1 dysfunction is associated with brain disorders are also poorly understood. In the current study, we found that Notch1 was highly expressed in the adult-born immature neurons in the hippocampal dentate gyrus. Using a genetic approach to selectively ablate Notch1 signaling in late immature precursors in the postnatal hippocampus by cross-breeding doublecortin (DCX)+ neuron-specific proopiomelanocortin (POMC)-α Cre mice with floxed Notch1 mice, we demonstrated a previously unreported pivotal role of Notch1 signaling in survival and function of adult newborn neurons in the dentate gyrus. Moreover, behavioral and functional studies demonstrated that POMC-Notch1-/- mutant mice showed anxiety and depressive-like behavior with impaired synaptic transmission properties in the dentate gyrus. Finally, our mechanistic study showed significantly compromised phosphorylation of cAMP response element-binding protein (CREB) in Notch1 mutants, suggesting that the dysfunction of Notch1 mutants is associated with the disrupted pCREB signaling in postnatally generated immature neurons in the dentate gyrus.-Feng, S., Shi, T., Qiu, J., Yang, H., Wu, Y., Zhou, W., Wang, W., Wu, H. Notch1 deficiency in postnatal neural progenitor cells in the dentate gyrus leads to emotional and cognitive impairment.


Assuntos
Ansiedade , Disfunção Cognitiva/metabolismo , Giro Denteado/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Receptor Notch1/metabolismo , Animais , Ansiedade/genética , Disfunção Cognitiva/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína Duplacortina , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Camundongos Knockout , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Receptor Notch1/deficiência , Transdução de Sinais/fisiologia
11.
J Pathol ; 242(4): 448-462, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28497579

RESUMO

In stratified epithelia such as the epidermis, homeostasis is maintained by the proliferation of cells in the lower epithelial layers and the concomitant loss of differentiated cells from the epithelial surface. These differentiating keratinocytes progressively stratify and form a self-regenerating multi-layered barrier that protects the underlying dermis. In such tissue, the continual loss and replacement of differentiated cells also limits the accumulation of oncogenic mutations within the tissue. Inactivating mutations in key driver genes, such as TP53 and NOTCH1, reduce the proportion of differentiating cells allowing for the long-term persistence of expanding mutant clones in the tissue. Here we show that through the expression of E6, HPV-16 prevents the early fate commitment of human keratinocytes towards differentiation and confers a strong growth advantage to human keratinocytes. When E6 is expressed either alone or with E7, it promotes keratinocyte proliferation at high cell densities, through the combined inactivation of p53 and Notch1. In organotypic raft culture, the activity of E6 is restricted to the basal layer of the epithelium and is enhanced during the progression from productive to abortive or transforming HPV-16 infection. Consistent with this, the expression of p53 and cleaved Notch1 becomes progressively more disrupted, and is associated with increased basal cell density and reduced commitment to differentiation. The expression of cleaved Notch1 is similarly disrupted also in HPV-16-positive cervical lesions, depending on neoplastic grade. When taken together, these data depict an important role of high-risk E6 in promoting the persistence of infected keratinocytes in the basal and parabasal layers through the inactivation of gene products that are commonly mutated in non-HPV-associated neoplastic squamous epithelia. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Papillomavirus Humano 16/fisiologia , Proteínas Oncogênicas Virais/fisiologia , Infecções por Papillomavirus/metabolismo , Receptor Notch1/metabolismo , Proteínas Repressoras/fisiologia , Neoplasias do Colo do Útero/virologia , Diferenciação Celular/fisiologia , Divisão Celular , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Transformação Celular Viral/fisiologia , Progressão da Doença , Feminino , Humanos , Queratinócitos/patologia , Queratinócitos/virologia , Gradação de Tumores , Infecções por Papillomavirus/patologia , RNA Mensageiro/genética , Receptor Notch1/deficiência , Receptor Notch1/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
12.
Lab Invest ; 96(9): 972-80, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27428080

RESUMO

Notch signaling pathways have recently been implicated in the pathogenesis of metabolic diseases. However, the role of hepatic Notch signaling in glucose and lipid metabolism remains unclear and needs further investigation as it might be a candidate therapeutic target in metabolic diseases such as nonalcoholic steatohepatitis (NASH) and nonalcoholic fatty liver disease (NAFLD). We used hepatocyte-specific Notch1 knockout (KO) mice and liver biopsies from NASH and NAFLD patients to analyze the role of Notch1 in glucose and lipid metabolism. Hepatocyte-specific Notch1 KO mice were fed with a high fat diet (HFD) or a regular diet (RD). We assessed the metabolic phenotype, glucose and insulin tolerance tests, and liver histology. Hepatic mRNA expression was profiled by Affymetrix Mouse Gene arrays and validated by quantitative reverse transcription PCR (qPCR). Akt phosphorylation was visualized by immunoblotting. Gene expression was analyzed in liver biopsies from NASH, NAFLD, and control patients by qPCR. We found that Notch1 KO mice had elevated fasting glucose. Gene expression analysis showed an upregulation of glucose-6-phosphatase, involved in the final step of gluconeogenesis and glucose release from glycogenolysis, and perilipin-5, a regulator of hepatic lipid accumulation. When fed with an HFD KO mice developed overt diabetes and hepatic steatosis. Akt was highly phosphorylated in KO animals and the Foxo1 target gene expression was altered. Accordingly, a reduction in Notch1 and increase in glucose-6-phosphatase and perilipin-5 expression was observed in liver biopsies from NAFLD/NASH compared with controls. Notch1 is a regulator of hepatic glucose and lipid homeostasis. Hepatic impairment of Notch1 expression may be involved in the pathogenesis of human NAFLD/NASH.


Assuntos
Diabetes Mellitus/genética , Fígado Gorduroso/genética , Predisposição Genética para Doença/genética , Glucose-6-Fosfatase/genética , Perilipina-1/genética , Receptor Notch1/genética , Animais , Diabetes Mellitus/etiologia , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/etiologia , Perfilação da Expressão Gênica/métodos , Hepatócitos/metabolismo , Humanos , Immunoblotting , Fígado/metabolismo , Fígado/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Notch1/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
13.
Circ Res ; 119(4): 519-31, 2016 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-27354212

RESUMO

RATIONALE: Endothelial Notch signaling is critical for early vascular development and survival. Yet, previously described mice lacking endothelial a disintegrin and metalloproteinase 10 (ADAM10), a key regulator of Notch signaling, survived into adulthood with organ-specific vascular defects. These findings raised questions about whether these vascular defects were related to Notch signaling or other functions of ADAM10. OBJECTIVE: The aims of the study are to determine whether compensatory or redundant functions of ADAM17 in Notch signaling can explain the survival of Adam10ΔEC mice, explore the contribution of different Tie2-Cre transgenes to the differences in survival, and establish whether the Adam10ΔEC vascular phenotypes can be recapitulated by inactivation of Notch receptors in endothelial cells. METHODS AND RESULTS: Mice lacking ADAM10 and ADAM17 in endothelial cells (Adam10/Adam17ΔEC), which survived postnatally with organ-specific vascular defects, resembled Adam10ΔEC mice. In contrast, Adam10ΔEC mice generated with the Tie2Cre transgene previously used to inactivate endothelial Notch (Adam10ΔEC(Flv)) died by E10.5. Quantitative polymerase chain reaction analysis demonstrated that Cre-mediated recombination occurs earlier in Adam10ΔEC(Flv) mice than in the previously described Adam10ΔEC mice. Finally, mice lacking endothelial Notch1 (Notch1ΔEC) share some organ-specific vascular defects with Adam10ΔEC mice, whereas Notch4(-/-) mice lacking endothelial Notch1 (Notch1ΔEC/Notch4(-/-)) had defects in all vascular beds affected in Adam10ΔEC mice. CONCLUSIONS: Our results argue against a major role for ADAM17 in endothelial Notch signaling and clarify the difference in phenotypes of previously described mice lacking ADAM10 or Notch in endothelial cells. Most notably, these findings uncover new roles for Notch signaling in the development of organ-specific vascular beds.


Assuntos
Proteína ADAM10/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Circulação Sanguínea/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptor Notch1/fisiologia , Receptores Notch/fisiologia , Fluxo Sanguíneo Regional/fisiologia , Transdução de Sinais/fisiologia , Proteína ADAM10/deficiência , Secretases da Proteína Precursora do Amiloide/deficiência , Animais , Células Endoteliais/fisiologia , Feminino , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Gravidez , Proteínas Proto-Oncogênicas/deficiência , Receptor Notch1/deficiência , Receptor Notch4 , Receptores Notch/deficiência
14.
Sci Rep ; 6: 19377, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26786165

RESUMO

Notch signaling pathways modulate various cellular processes, including cell proliferation, differentiation, adhesion, and communication. Recent studies have demonstrated that Notch1 signaling also regulates hepatic glucose production and lipid synthesis. However, the effect of Notch1 signaling on hepatic lipid oxidation has not yet been directly investigated. To define the function of Notch1 signaling in hepatic lipid metabolism, wild type mice and Notch1 deficient antisense transgenic (NAS) mice were fed a high-fat diet. High-fat diet -fed NAS mice exhibited a marked reduction in hepatic triacylglycerol accumulation compared with wild type obese mice. The improved fatty liver was associated with an increased expression of hepatic genes involved in fatty acid oxidation. However, lipogenic genes were not differentially expressed in the NAS liver, suggesting lipolytic-specific regulatory effects by Notch1 signaling. Expression of fatty acid oxidative genes and the rate of fatty acid oxidation were also increased by inhibition of Notch1 signaling in HepG2 cells. In addition, similar regulatory effects on lipid accumulation were observed in adipocytes. Taken together, these data show that inhibition of Notch1 signaling can regulate the expression of fatty acid oxidation genes and may provide therapeutic strategies in obesity-induced hepatic steatosis.


Assuntos
Ácidos Graxos/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Oxirredução , Receptor Notch1/deficiência , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Linhagem Celular , Dieta/efeitos adversos , Fígado Gorduroso/patologia , Técnicas de Silenciamento de Genes , Humanos , Resistência à Insulina/genética , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos , Obesidade/genética , Obesidade/metabolismo , Oxirredução/efeitos dos fármacos , Estresse Oxidativo , Interferência de RNA , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Hippocampus ; 26(5): 589-600, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26482652

RESUMO

It is well known that the neuronal effects of vascular endothelial growth factor (VEGF) include modulating learning and memory, plasticity of mature neurons, and synaptic transmission in addition to neurogenesis. However, there is conflicting evidence particularly of its role in the regulation of excitatory synaptic activity. In this study, application of the patch-clamp technique revealed that lower doses (10 and 50 ng/mL) of VEGF enhanced excitatory neurotransmission in hippocampal slices of mice through both presynaptic and postsynaptic mechanisms. However, the effects were reversed by higher doses of VEGF (>100 ng/mL), which inhibited excitatory neurotransmission via a presynaptic mechanism. These competing, concentration-dependent effects of VEGF suggested that different pathways were involved. The involvement of the Notch1 receptor was tested in the modulation of VEGF on synaptic activity by using heterozygous Notch1(+/-) mice. Notch1 knockdown did not influence the inhibitory effect of high VEGF doses (200 ng/mL) but reduced the enhancement effects of low concentration of VEGF (50 ng/mL) at the postsynaptic level, which might be due to the decreased level of VEGF receptor. The results indicate that the Notch1 receptor plays a role in VEGF-induced modulation of synaptic activity, which provides new insights into a complex VEGF/Notch signaling cross-talk. These findings set the groundwork for understanding new mechanisms of Notch signaling and the neurotrophic effects of VEGF, which is beneficial to develop new therapeutic targets to the VEGF/Notch axis and improve current treatments for neural diseases.


Assuntos
Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Receptor Notch1/deficiência , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Animais Recém-Nascidos , Fenômenos Biofísicos/efeitos dos fármacos , Fenômenos Biofísicos/genética , Relação Dose-Resposta a Droga , Estimulação Elétrica , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/genética , Neurônios/fisiologia , Receptor Notch1/genética , Estatísticas não Paramétricas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
16.
Nat Cell Biol ; 18(2): 168-80, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26689676

RESUMO

Chronic inflammation is associated with a variety of pathological conditions in epithelial tissues, including cancer, metaplasia and aberrant wound healing. In relation to this, a significant body of evidence suggests that aberration of epithelial stem and progenitor cell function is a contributing factor in inflammation-related disease, although the underlying cellular and molecular mechanisms remain to be fully elucidated. In this study, we have delineated the effect of chronic inflammation on epithelial stem/progenitor cells using the corneal epithelium as a model tissue. Using a combination of mouse genetics, pharmacological approaches and in vitro assays, we demonstrate that chronic inflammation elicits aberrant mechanotransduction in the regenerating corneal epithelium. As a consequence, a YAP-TAZ/ß-catenin cascade is triggered, resulting in the induction of epidermal differentiation on the ocular surface. Collectively, the results of this study demonstrate that chronic inflammation and mechanotransduction are linked and act to elicit pathological responses in regenerating epithelia.


Assuntos
Diferenciação Celular , Lesões da Córnea/metabolismo , Células Epiteliais/metabolismo , Epitélio Corneano/metabolismo , Ceratite/metabolismo , Mecanotransdução Celular , Regeneração , Células-Tronco/metabolismo , Aciltransferases , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Administração Oftálmica , Animais , Anti-Inflamatórios/administração & dosagem , Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Doença Crônica , Lesões da Córnea/genética , Lesões da Córnea/patologia , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/lesões , Epitélio Corneano/patologia , Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Ceratite/genética , Ceratite/patologia , Ceratite/prevenção & controle , Mecanotransdução Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptor Notch1/deficiência , Receptor Notch1/genética , Regeneração/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Suínos , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Via de Sinalização Wnt , Proteínas de Sinalização YAP , beta Catenina/genética , beta Catenina/metabolismo
17.
Sci Rep ; 5: 16449, 2015 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-26563570

RESUMO

Pericytes regulate vessel stability and pericyte dysfunction contributes to retinopathies, stroke, and cancer. Here we define Notch as a key regulator of pericyte function during angiogenesis. In Notch1(+/-); Notch3(-/-) mice, combined deficiency of Notch1 and Notch3 altered pericyte interaction with the endothelium and reduced pericyte coverage of the retinal vasculature. Notch1 and Notch3 were shown to cooperate to promote proper vascular basement membrane formation and contribute to endothelial cell quiescence. Accordingly, loss of pericyte function due to Notch deficiency exacerbates endothelial cell activation caused by Notch1 haploinsufficiency. Mice mutant for Notch1 and Notch3 develop arteriovenous malformations and display hallmarks of the ischemic stroke disease CADASIL. Thus, Notch deficiency compromises pericyte function and contributes to vascular pathologies.


Assuntos
Malformações Arteriovenosas/genética , CADASIL/genética , Pericitos/metabolismo , Receptor Notch1/genética , Receptores Notch/genética , Animais , Malformações Arteriovenosas/metabolismo , Western Blotting , CADASIL/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/ultraestrutura , Expressão Gênica , Células HEK293 , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Pericitos/patologia , Pericitos/ultraestrutura , Receptor Notch1/deficiência , Receptor Notch3 , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores Notch/deficiência , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Vasos Retinianos/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
18.
Nat Immunol ; 16(11): 1162-73, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26437242

RESUMO

Receptors of the Notch family direct the differentiation of helper T cell subsets, but their influence on regulatory T cell (T(reg) cell) responses is obscure. We found here that lineage-specific deletion of components of the Notch pathway enhanced T(reg) cell-mediated suppression of type 1 helper T cell (T(H)1 cell) responses and protected against their T(H)1 skewing and apoptosis. In contrast, expression in T(reg) cells of a gain-of-function transgene encoding the Notch1 intracellular domain resulted in lymphoproliferation, exacerbated T(H)1 responses and autoimmunity. Cell-intrinsic canonical Notch signaling impaired T(reg) cell fitness and promoted the acquisition by T(reg) cells of a T(H)1 cell-like phenotype, whereas non-canonical Notch signaling dependent on the adaptor Rictor activated the kinase AKT-transcription factor Foxo1 axis and impaired the epigenetic stability of Foxp3. Our findings establish a critical role for Notch signaling in controlling peripheral T(reg) cell function.


Assuntos
Tolerância Periférica , Receptor Notch1/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Epigênese Genética , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Proteína Companheira de mTOR Insensível à Rapamicina , Receptor Notch1/deficiência , Receptor Notch1/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/imunologia , Células Th1/imunologia , Transcriptoma
19.
Am J Pathol ; 185(11): 3066-75, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26358219

RESUMO

Notch signaling pathway is involved in the regulation of cell fate, differentiation, proliferation, and apoptosis in development and disease. Previous studies suggest the importance of Notch1 in myofibroblast differentiation in lung alveogenesis and fibrosis. However, direct in vivo evidence of Notch1-mediated myofibroblast differentiation is lacking. In this study, we examined the effects of conditional mesenchymal-specific deletion of Notch1 on pulmonary fibrosis. Crossing of mice bearing the floxed Notch1 gene with α2(I) collagen enhancer-Cre-ER(T)-bearing mice successfully generated progeny with a conditional knockout (CKO) of Notch1 in collagen I-expressing (mesenchymal) cells on treatment with tamoxifen (Notch1 CKO). Because Notch signaling is known to be activated in the bleomycin model of pulmonary fibrosis, control and Notch1 CKO mice were analyzed for their responses to bleomycin treatment. The results showed significant attenuation of pulmonary fibrosis in CKO relative to control mice, as examined by collagen deposition, myofibroblast differentiation, and histopathology. However, there were no significant differences in inflammatory or immune cell influx between bleomycin-treated CKO and control mouse lungs. Analysis of isolated lung fibroblasts confirmed absence of Notch1 expression in cells from CKO mice, which contained fewer myofibroblasts and significantly diminished collagen I expression relative to those from control mice. These findings revealed an essential role for Notch1-mediated myofibroblast differentiation in the pathogenesis of pulmonary fibrosis.


Assuntos
Bleomicina/efeitos adversos , Colágeno/metabolismo , Fibrose Pulmonar/patologia , Receptor Notch1/deficiência , Transdução de Sinais , Animais , Apoptose , Diferenciação Celular , Colágeno/genética , Fibroblastos/metabolismo , Fibroblastos/patologia , Pulmão/metabolismo , Pulmão/patologia , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fibrose Pulmonar/induzido quimicamente , Receptor Notch1/genética , Deleção de Sequência
20.
Cancer Res ; 75(18): 3958-3969, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26294213

RESUMO

Viral oncogene expression is insufficient for neoplastic transformation of human cells, so human papillomavirus (HPV)-associated cancers will also rely upon mutations in cellular oncogenes and tumor suppressors. However, it has been difficult so far to distinguish incidental mutations without phenotypic impact from causal mutations that drive the development of HPV-associated cancers. In this study, we addressed this issue by conducting a functional screen for genes that facilitate the formation of HPV E6/E7-induced squamous cell cancers in mice using a transposon-mediated insertional mutagenesis protocol. Overall, we identified 39 candidate driver genes, including Notch1, which unexpectedly was scored by gain- or loss-of-function mutations that were capable of promoting squamous cell carcinogenesis. Autochthonous HPV-positive oral tumors possessing an activated Notch1 allele exhibited high rates of cell proliferation and tumor growth. Conversely, Notch1 loss could accelerate the growth of invasive tumors in a manner associated with increased expression of matrix metalloproteinases and other proinvasive genes. HPV oncogenes clearly cooperated with loss of Notch1, insofar as its haploinsufficiency accelerated tumor growth only in HPV-positive tumors. In clinical specimens of various human cancers, there was a consistent pattern of NOTCH1 expression that correlated with invasive character, in support of our observations in mice. Although Notch1 acts as a tumor suppressor in mouse skin, we found that oncogenes enabling any perturbation in Notch1 expression promoted tumor growth, albeit via distinct pathways. Our findings suggest caution in interpreting the meaning of putative driver gene mutations in cancer, and therefore therapeutic efforts to target them, given the significant contextual differences in which such mutations may arise, including in virus-associated tumors.


Assuntos
Transformação Celular Neoplásica , Transformação Celular Viral , Cocarcinogênese , Neoplasias Bucais/genética , Oncogenes , Papillomaviridae/patogenicidade , Receptor Notch1/fisiologia , Infecções Tumorais por Vírus/fisiopatologia , 4-Nitroquinolina-1-Óxido/toxicidade , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/virologia , Carcinógenos , Carcinoma Verrucoso/patologia , Carcinoma Verrucoso/virologia , Elementos de DNA Transponíveis , Progressão da Doença , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias Bucais/virologia , Mutagênese Insercional , Invasividade Neoplásica , Papiloma/induzido quimicamente , Papiloma/patologia , Papiloma/virologia , Receptor Notch1/deficiência , Receptor Notch1/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Organismos Livres de Patógenos Específicos , Tamoxifeno/farmacologia , Acetato de Tetradecanoilforbol/toxicidade , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA