Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Am Soc Nephrol ; 25(12): 2717-29, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24854275

RESUMO

Ischemia-reperfusion (I/R) injury of the kidney is a major cause of AKI. MicroRNAs (miRs) are powerful regulators of various diseases. We investigated the role of apoptosis-associated miR-24 in renal I/R injury. miR-24 was upregulated in the kidney after I/R injury of mice and in patients after kidney transplantation. Cell-sorting experiments revealed a specific miR-24 enrichment in renal endothelial and tubular epithelial cells after I/R induction. In vitro, anoxia/hypoxia induced an enrichment of miR-24 in endothelial and tubular epithelial cells. Transient overexpression of miR-24 alone induced apoptosis and altered functional parameters in these cells, whereas silencing of miR-24 ameliorated apoptotic responses and rescued functional parameters in hypoxic conditions. miR-24 effects were mediated through regulation of H2A histone family, member X, and heme oxygenase 1, which were experimentally validated as direct miR-24 targets through luciferase reporter assays. In vitro, adenoviral overexpression of miR-24 targets lacking miR-24 binding sites along with miR-24 precursors rescued various functional parameters in endothelial and tubular epithelial cells. In vivo, silencing of miR-24 in mice before I/R injury resulted in a significant improvement in survival and kidney function, a reduction of apoptosis, improved histologic tubular epithelial injury, and less infiltration of inflammatory cells. miR-24 also regulated heme oxygenase 1 and H2A histone family, member X, in vivo. Overall, these results indicate miR-24 promotes renal ischemic injury by stimulating apoptosis in endothelial and tubular epithelial cell. Therefore, miR-24 inhibition may be a promising future therapeutic option in the treatment of patients with ischemic AKI.


Assuntos
Túbulos Renais/metabolismo , Rim/metabolismo , Rim/patologia , MicroRNAs/antagonistas & inibidores , Traumatismo por Reperfusão/patologia , Adulto , Animais , Apoptose , Sítios de Ligação , Células Endoteliais/citologia , Endotélio/patologia , Células Epiteliais/metabolismo , Feminino , Inativação Gênica , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/metabolismo , Histonas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/metabolismo , Túbulos Renais/patologia , Masculino , Camundongos , MicroRNAs/genética , Pessoa de Meia-Idade , Receptores de Lisoesfingolipídeo/metabolismo , Receptores de Esfingosina-1-Fosfato
3.
Cancer Cell ; 23(6): 784-95, 2013 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-23727022

RESUMO

The incidence of cholangiocellular carcinoma (CCC) is increasing worldwide. Using a transgenic mouse model, we found that expression of the intracellular domain of Notch 1 (NICD) in mouse livers results in the formation of intrahepatic CCCs. These tumors display features of bipotential hepatic progenitor cells, indicating that intrahepatic CCC can originate from this cell type. We show that human and mouse CCCs are characterized by high expression of the cyclin E protein and identified the cyclin E gene as a direct transcriptional target of the Notch signaling pathway. Intriguingly, blocking γ-secretase activity in human CCC xenotransplants results in downregulation of cyclin E expression, induction of apoptosis, and tumor remission in vivo.


Assuntos
Colangiocarcinoma/genética , Regulação Neoplásica da Expressão Gênica , Receptor Notch1/genética , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Apoptose/genética , Proliferação de Células , Colangiocarcinoma/patologia , Ciclina E/genética , Ciclina E/metabolismo , Regulação para Baixo , Hepatócitos/citologia , Humanos , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Receptor Notch1/química , Receptor Notch1/metabolismo , Transdução de Sinais , Transplante Heterólogo
4.
J Am Soc Nephrol ; 22(10): 1887-96, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21841063

RESUMO

Renal ischemia-reperfusion contributes to reduced renal allograft survival. The peptide Bß(15-42), a breakdown product of fibrin, attenuates inflammation induced by ischemia-reperfusion in the heart by competitively blocking the binding of leukocytes to endothelial VE-cadherin, but whether it could improve outcomes in renal transplantation is unknown. Here, we tested the ability of Bß(15-42) to ameliorate the effects of renal ischemic injury during allogenic kidney transplantation in mice. In our renal transplantation model (C57BL/6 into BALB/c mice), treatment with Bß(15-42) at the time of allograft reperfusion resulted in significantly improved survival of recipients during the 28-day follow-up (60% versus 10%). Bß(15-42) treatment decreased leukocyte infiltration, expression of endothelial adhesion molecules, and proinflammatory cytokines. Treatment significantly attenuated allogenic T cell activation and reduced cellular rejection. Moreover, Bß(15-42) significantly reduced tubular epithelial damage and apoptosis, which we reproduced in vitro. These data suggest that Bß(15-42) may have therapeutic potential in transplant surgery by protecting grafts from ischemia-reperfusion injury.


Assuntos
Injúria Renal Aguda/prevenção & controle , Produtos de Degradação da Fibrina e do Fibrinogênio/administração & dosagem , Transplante de Rim/efeitos adversos , Fragmentos de Peptídeos/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Feminino , Transplante de Rim/imunologia , Túbulos Renais/citologia , Complexo Principal de Histocompatibilidade/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/efeitos dos fármacos , Transplante Homólogo
5.
Kidney Int ; 80(10): 1035-44, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21734641

RESUMO

Fibrinogen plays an important role in blood coagulation but its function extends far beyond blood clotting being involved in inflammation and repair. Besides these crucial functions it can also promote tissue fibrosis. To determine whether fibrinogen is involved in the development of renal tubulointerstitial fibrosis we utilized the profibrotic model of unilateral ureteral obstruction in fibrinogen-deficient mice. In the heterozygotes, obstruction was associated with a massive deposition of intrarenal fibrinogen. Fibrinogen deficiency provided significant protection from interstitial damage and tubular disruption, attenuated collagen accumulation, and greatly reduced de novo expression of α-smooth muscle actin in the obstructed kidney. While no differences were found in renal inflammatory cell infiltration, fibrinogen deficiency was associated with a significant reduction in interstitial cell proliferation, a hallmark of renal fibrosis. In vitro, fibrinogen directly stimulated renal fibroblast proliferation in a dose-dependent manner. This mitogenic effect of fibrinogen was mediated by at least three different cell surface receptors on renal fibroblasts: TLR2, TLR4, and ICAM-1. Thus, our study suggests that fibrinogen promotes renal fibrosis by triggering resident fibroblast proliferation.


Assuntos
Proliferação de Células , Fibrinogênio/metabolismo , Fibroblastos/metabolismo , Túbulos Renais/metabolismo , Obstrução Ureteral/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Fibrinogênio/genética , Fibroblastos/patologia , Fibrose , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Túbulos Renais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Interferência de RNA , Ratos , Fatores de Tempo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Transfecção , Obstrução Ureteral/genética , Obstrução Ureteral/patologia
6.
J Neurosci ; 29(48): 15073-82, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19955358

RESUMO

In contrast to mammals, the spinal cord of adult zebrafish has the capacity to reinitiate generation of motor neurons after a lesion. Here we show that genes involved in motor neuron development, i.e., the ventral morphogen sonic hedgehog a (shha), as well as the transcription factors nkx6.1 and pax6, together with a Tg(olig2:egfp) transgene, are expressed in the unlesioned spinal cord of adult zebrafish. Expression is found in ependymoradial glial cells lining the central canal in ventrodorsal positions that match expression domains of these genes in the developing neural tube. Specifically, Tg(olig2:egfp)(+) ependymoradial glial cells, the adult motor neuron progenitors (pMNs), coexpress Nkx6.1 and Pax6, thus defining an adult pMN-like zone. shha is expressed in distinct ventral ependymoradial glial cells. After a lesion, expression of all these genes is strongly increased, while relative spatial expression domains are maintained. In addition, expression of the hedgehog (hh) receptors patched1 and smoothened becomes detectable in ependymoradial glial cells including those of the pMN-like zone. Cyclopamine-induced knock down of hh signaling significantly reduces ventricular proliferation and motor neuron regeneration. Expression of indicator genes for the FGF and retinoic acid signaling pathways was also increased in the lesioned spinal cord. This suggests that a subclass of ependymoradial glial cells retain their identity as motor neuron progenitors into adulthood and are capable of reacting to a sonic hedgehog signal and potentially other developmental signals with motor neuron regeneration after a spinal lesion.


Assuntos
Polaridade Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas Hedgehog/fisiologia , Neurônios Motores/fisiologia , Regeneração Nervosa/fisiologia , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Polaridade Celular/genética , Proliferação de Células , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas Hedgehog/genética , Lisina/análogos & derivados , Lisina/metabolismo , Neurônios Motores/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Neuroglia/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , RNA Mensageiro/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/genética , Transdução de Sinais/genética , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Estatísticas não Paramétricas , Fatores de Transcrição/genética , Alcaloides de Veratrum/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Cell ; 137(6): 1124-35, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19524514

RESUMO

The Notch pathway is a highly conserved signaling system that controls a diversity of growth, differentiation, and patterning processes. In growing blood vessels, sprouting of endothelial tip cells is inhibited by Notch signaling, which is activated by binding of the Notch receptor to its ligand Delta-like 4 (Dll4). Here, we show that the Notch ligand Jagged1 is a potent proangiogenic regulator in mice that antagonizes Dll4-Notch signaling in cells expressing Fringe family glycosyltransferases. Upon glycosylation of Notch, Dll4-Notch signaling is enhanced, whereas Jagged1 has weak signaling capacity and competes with Dll4. Our findings establish that the equilibrium between two Notch ligands with distinct spatial expression patterns and opposing functional roles regulates angiogenesis, a mechanism that might also apply to other Notch-controlled biological processes.


Assuntos
Vasos Sanguíneos/embriologia , Proteínas de Ligação ao Cálcio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Neovascularização Fisiológica , Proteínas Adaptadoras de Transdução de Sinal , Animais , Vasos Sanguíneos/citologia , Proteínas de Ligação ao Cálcio/genética , Embrião de Mamíferos/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Feminino , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Mutação , Receptores Notch/metabolismo , Retina/embriologia , Proteínas Serrate-Jagged
8.
Blood ; 113(22): 5680-8, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19144989

RESUMO

Notch signaling has been shown to regulate various aspects of vascular development. However, a specific role of the ligand Delta-like 1 (DLL1) has not been shown thus far. Here, we demonstrate that during fetal development, DLL1 is an essential Notch ligand in the vascular endothelium of large arteries to activate Notch1 and maintain arterial identity. DLL1 was detected in fetal arterial endothelial cells beginning at embryonic day 13.5. While DLL4-mediated activation has been shown to suppress vascular endothelial growth factor (VEGF) pathway components in growing capillary beds, DLL1-Notch signaling was required for VEGF receptor expression in fetal arteries. In the absence of DLL1 function, VEGF receptor 2 (VEGFR2) and its coreceptor, neuropilin-1 (NRP1), were down-regulatedin mutant arteries, which was followed by up-regulation of chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII), a repressor of arterial differentiation and Nrp1 expression in veins. Consistent with a positive modulation of the VEGF pathway by DLL1, the Nrp1 promoter contains several recombinant signal-binding protein 1 for J kappa (RBPJkappa)-binding sites and was responsive to Notch activity in cell culture. Our results establish DLL1 as a critical endothelial Notch ligand required for maintaining arterial identity during mouse fetal development and suggest context-dependent interrelations of the VEGFA and Notch signaling pathways.


Assuntos
Artérias/embriologia , Diferenciação Celular/genética , Células Endoteliais/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Receptor Notch1/metabolismo , Animais , Artérias/metabolismo , Células CHO , Proteínas de Ligação ao Cálcio , Cricetinae , Cricetulus , Embrião de Mamíferos , Células Endoteliais/metabolismo , Feto/irrigação sanguínea , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Receptor Notch1/fisiologia , Transdução de Sinais/genética
9.
J Neurosci ; 28(34): 8510-6, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18716209

RESUMO

The mammalian spinal cord does not regenerate motor neurons that are lost as a result of injury or disease. Here we demonstrate that adult zebrafish, which show functional spinal cord regeneration, are capable of motor neuron regeneration. After a spinal lesion, the ventricular zone shows a widespread increase in proliferation, including slowly proliferating olig2-positive (olig2+) ependymo-radial glial progenitor cells. Lineage tracing in olig2:green fluorescent protein transgenic fish indicates that these cells switch from a gliogenic phenotype to motor neuron production. Numbers of undifferentiated small HB9+ and islet-1+ motor neurons, which are double labeled with the proliferation marker 5-bromo-2-deoxyuridine (BrdU), are transiently strongly increased in the lesioned spinal cord. Large differentiated motor neurons, which are lost after a lesion, reappear at 6-8 weeks after lesion, and we detected ChAT+/BrdU+ motor neurons that were covered by contacts immunopositive for the synaptic marker SV2. These observations suggest that, after a lesion, plasticity of olig2+ progenitor cells may allow them to generate motor neurons, some of which exhibit markers for terminal differentiation and integration into the existing adult spinal circuitry.


Assuntos
Neurônios Motores , Regeneração Nervosa , Traumatismos da Medula Espinal/fisiopatologia , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bromodesoxiuridina , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM , Microscopia Eletrônica , Neurônios Motores/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Fator de Transcrição 2 de Oligodendrócitos , Fenótipo , Proteínas Recombinantes de Fusão/genética , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/patologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
Cancer Cell ; 14(1): 23-35, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18598941

RESUMO

A reduction in the cellular levels of the cyclin kinase inhibitor p27(kip1) is frequently found in many human cancers and correlates directly with patient prognosis. In this work, we identify argyrin A, a cyclical peptide derived from the myxobacterium Archangium gephyra, as a potent antitumoral drug. All antitumoral activities of argyrin A depend on the prevention of p27(kip1) destruction, as loss of p27(kip1) expression confers resistance to this compound. We find that argyrin A exerts its effects through a potent inhibition of the proteasome. By comparing the cellular responses exerted by argyrin A with siRNA-mediated knockdown of proteasomal subunits, we find that the biological effects of proteasome inhibition per se depend on the expression of p27(kip1).


Assuntos
Inibidores da Angiogênese/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias/tratamento farmacológico , Peptídeos Cíclicos/farmacologia , Inibidores de Proteassoma , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Ácidos Borônicos/farmacologia , Bortezomib , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27 , Relação Dose-Resposta a Droga , Células HCT116 , Células HeLa , Humanos , Camundongos , Camundongos Nus , Necrose , Neoplasias/irrigação sanguínea , Neoplasias/enzimologia , Neoplasias/patologia , Neovascularização Patológica/enzimologia , Neovascularização Patológica/prevenção & controle , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirazinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Transfecção , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Circ Res ; 100(3): 363-71, 2007 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-17234965

RESUMO

Growth of functional arteries is essential for the restoration of blood flow to ischemic organs. Notch signaling regulates arterial differentiation upstream of ephrin-B2 during embryonic development, but its role during postnatal arteriogenesis is unknown. Here, we identify the Notch ligand Delta-like 1 (Dll1) as an essential regulator of postnatal arteriogenesis. Dll1 expression was specifically detected in arterial endothelial cells, but not in venous endothelial cells or capillaries. During ischemia-induced arteriogenesis endothelial Dll1 expression was strongly induced, Notch signaling activated and ephrin-B2 upregulated, whereas perivascular cells expressed proangiogenic vascular endothelial growth factor, and the ephrin-B2 activator EphB4. In heterozygous Dll1 mutant mice endothelial Notch activation and ephrin-B2 induction after hindlimb ischemia were absent, arterial collateral growth was abrogated and recovery of blood flow was severely impaired, but perivascular vascular endothelial growth factor and EphB4 expression was unaltered. In vitro, angiogenic growth factors synergistically activated Notch signaling by induction of Dll1, which was necessary and sufficient to regulate ephrin-B2 expression and to induce ephrin-B2 and EphB4-dependent branching morphogenesis in human arterial EC. Thus, Dll1-mediated Notch activation regulates ephrin-B2 expression and postnatal arteriogenesis.


Assuntos
Artérias/citologia , Endotélio Vascular/citologia , Regulação da Expressão Gênica/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Isquemia/fisiopatologia , Proteínas de Membrana/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores Notch/fisiologia , Animais , Aorta/citologia , Artérias/química , Artérias/crescimento & desenvolvimento , Proteínas de Ligação ao Cálcio , Capilares/química , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Circulação Colateral/fisiologia , Constrição , Meios de Cultura Livres de Soro , Células Endoteliais/metabolismo , Inativação Gênica , Membro Posterior/irrigação sanguínea , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Isquemia/etiologia , Isquemia/genética , Camundongos , Camundongos Transgênicos , Morfogênese/genética , Morfogênese/fisiologia , Neovascularização Fisiológica/genética , Especificidade de Órgãos , RNA Interferente Pequeno/farmacologia , Receptor EphB2/biossíntese , Receptor EphB2/genética , Receptor EphB2/fisiologia , Receptor EphB4/biossíntese , Receptor EphB4/genética , Receptor EphB4/fisiologia , Veias/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA