Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 31(4): 842-50, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21273561

RESUMO

OBJECTIVE: Prokineticins are potent angiogenic hormones that use 2 receptors, prokineticin receptor-1 (PKR1) and PKR2, with important therapeutic use in anticancer therapy. Observations of cardiac and renal toxicity in cancer patients treated with antiangiogenic compounds led us to explore how PKR1 signaling functioned in heart and kidney in vivo. METHODS AND RESULTS: We generated mice with a conditional disruption of the PKR1 gene. We observed that PKR1 loss led to cardiomegaly, severe interstitial fibrosis, and cardiac dysfunction under stress conditions, accompanied by renal tubular dilation, reduced glomerular capillaries, urinary phosphate excretion, and proteinuria at later ages. Abnormal mitochondria and increased apoptosis were evident in both organs. Perturbation of capillary angiogenesis in both organs was restored at the adult stage potentially via upregulation of hypoxia-inducible factor-1 and proangiogenic factors. Compensatory mechanism could not revoke the epicardial and glomerular capillary networks, because of increased apoptosis and reduced progenitor cell numbers, consistent with an endogenous role of PKR1 signaling in stimulating epicardin+ progenitor cell proliferation and differentiation. CONCLUSIONS: Here, we showed for the first time that the loss of PKR1 causes renal and cardiac structural and functional changes because of deficits in survival signaling, mitochondrial, and progenitor cell functions in found both organs.


Assuntos
Inativação Gênica , Cardiopatias/genética , Nefropatias/genética , Rim/metabolismo , Miocárdio/metabolismo , Receptores Acoplados a Proteínas G/genética , Envelhecimento , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Predisposição Genética para Doença , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Rim/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Nefropatias/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Miocárdio/patologia , Neovascularização Fisiológica , Fenótipo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/patologia , Função Ventricular Esquerda
2.
Am J Physiol Heart Circ Physiol ; 298(3): H844-52, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20023120

RESUMO

Prokineticins are secreted peptides that activate two G protein-coupled receptors: PKR1 and PKR2. Prokineticins induce angiogenesis and fenestration, but the cognate receptors involved in these functions are unknown. We hypothesized a role for prokineticin receptor signaling pathways and expression profiles in determining the selective effects of prokineticins on coronary endothelial cells (H5V). Activation of the PKR1/MAPK/Akt signaling pathway stimulates proliferation, migration, and angiogenesis in H5V cells, in which PKR1 predominates over PKR2. PKR1 was colocalized with Galpha(11) and was internalized following the stimulation of these cells with prokineticin-2. Knock down of PKR1 or Galpha(11) expression in H5V cells effectively inhibited prokineticin-2-induced vessel formation and MAPK/Akt activation, indicating a role for PKR1/Galpha(11) in this process. However, in conditions in which PKR2 predominated over PKR1, these cells displayed a fenestrated endothelial cell phenotype. H5V cells overexpressing PKR2 displayed large numbers of multivesicular bodies and caveolar clusters and a disruption of the distribution of zonula occluden-1 tight junction protein. Prokineticin-2 induced the colocalization of PKR2 with Galpha(12), and activated Galpha(12), which bound to zonula occluden-1 to trigger the degradation of this protein in these cells. Prokineticin-2 induced the formation of vessel-like structures by human aortic endothelial cells expressing only PKR1, and disorganized the tight junctions in human hepatic sinusoidal endothelial cells expressing only PKR2, confirming the divergent roles of these receptors. Our findings show the functional characteristics of coronary endothelial cells depend on the expression of PKR1 and PKR2 levels and the divergent signaling pathways used by these receptors.


Assuntos
Endotélio Vascular/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Peptídeos/fisiologia , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Endotélio Vascular/citologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Modelos Animais , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Transdução de Sinais/fisiologia , Proteína da Zônula de Oclusão-1
3.
Arterioscler Thromb Vasc Biol ; 28(5): 841-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18421008

RESUMO

OBJECTIVE: Identification of novel factors that contribute to myocardial repair and collateral vessel growth hold promise for treatment of heart diseases. We have shown that transient prokineticin receptor-1 (PKR1) gene transfer protects the heart against myocardial infarction in a mouse model. Here, we investigated the role of excessive PKR1 signaling in heart. METHODS AND RESULTS: Transgenic mice overexpressing PKR1 in cardiomyocytes displayed no spontaneous abnormalities in cardiomyocytes but showed an increased number of epicardial-derived progenitor cells (EPDCs), capillary density, and coronary arterioles. Coculturing EPDCs with H9c2 cardiomyoblasts overexpressing PKR1 promotes EPDC differentiation into endothelial and smooth muscle cells, mimicking our transgenic model. Overexpressing PKR1 in H9c2 cardiomyoblasts or in transgenic hearts upregulated prokineticin-2 levels. Exogenous prokineticin-2 induces significant outgrowth from neonatal and adult epicardial explants, promoting EPDC differentiation. These prokineticin-2 effects were abolished in cardiac explants from mice with PKR1-null mutation. Reduced capillary density and prokineticin-2 levels in PKR1-null mutant hearts supports the hypothesis of an autocrine/paracrine loop between PKR1 and prokineticin-2. CONCLUSIONS: Cardiomyocyte-PKR1 signaling upregulates its own ligand prokineticin-2 that acts as a paracrine factor, triggering EPDCs proliferation/differentiation. This study provides a novel insight for possible therapeutic strategies aiming at restoring pluripotency of adult EPDCs to promote neovasculogenesis by induction of cardiomyocyte PKR1 signaling.


Assuntos
Diferenciação Celular/fisiologia , Neovascularização Fisiológica/fisiologia , Pericárdio/patologia , Proteína Quinase C/metabolismo , Células-Tronco/patologia , Animais , Movimento Celular/fisiologia , Proliferação de Células , Técnicas de Cocultura , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Pericárdio/metabolismo , Proteína Quinase C/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Regulação para Cima
4.
J Am Heart Assoc ; 2(5): e000411, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-24152983

RESUMO

BACKGROUND: Reciprocal relationships between endothelial dysfunction and insulin resistance result in a vicious cycle of cardiovascular, renal, and metabolic disorders. The mechanisms underlying these impairments are unclear. The peptide hormones prokineticins exert their angiogenic function via prokineticin receptor-1 (PKR1). We explored the extent to which endothelial PKR1 contributes to expansion of capillary network and the transcapillary passage of insulin into the heart, kidney, and adipose tissues, regulating organ functions and metabolism in a specific mice model. METHODS AND RESULTS: By combining cellular studies and studies in endothelium-specific loss-of-function mouse model (ec-PKR1-/-), we showed that a genetically induced PKR1 loss in the endothelial cells causes the impaired capillary formation and transendothelial insulin delivery, leading to insulin resistance and cardiovascular and renal disorders. Impaired insulin delivery in endothelial cells accompanied with defective expression and activation of endothelial nitric oxide synthase in the ec-PKR1-/- aorta, consequently diminishing endothelium-dependent relaxation. Despite having a lean body phenotype, ec-PKR1-/- mice exhibited polyphagia, polydipsia, polyurinemia, and hyperinsulinemia, which are reminiscent of human lipodystrophy. High plasma free fatty acid levels and low leptin levels further contribute to the development of insulin resistance at the later age. Peripheral insulin resistance and ectopic lipid accumulation in mutant skeletal muscle, heart, and kidneys were accompanied by impaired insulin-mediated Akt signaling in these organs. The ec-PKR1-/- mice displayed myocardial fibrosis, low levels of capillary formation, and high rates of apoptosis, leading to diastolic dysfunction. Compact fibrotic glomeruli and high levels of phosphate excretion were found in mutant kidneys. PKR1 restoration in ec-PKR1-/- mice reversed the decrease in capillary recruitment and insulin uptake and improved heart and kidney function and insulin resistance. CONCLUSIONS: We show a novel role for endothelial PKR1 signaling in cardiac, renal, and metabolic functions by regulating transendothelial insulin uptake and endothelial cell proliferation. Targeting endothelial PKR1 may serve as a therapeutic strategy for ameliorating these disorders.


Assuntos
Capilares/crescimento & desenvolvimento , Fenômenos Fisiológicos Cardiovasculares , Endotélio Vascular/metabolismo , Coração/fisiologia , Resistência à Insulina/fisiologia , Insulina/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Animais , Proliferação de Células , Endotélio Vascular/citologia , Masculino , Camundongos , Camundongos Transgênicos
5.
PLoS One ; 6(10): e25302, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22065986

RESUMO

BACKGROUND: Despite its effectiveness in the treatment of various cancers, the use of doxorubicin is limited by a potentially fatal cardiomyopathy. Prevention of this cardiotoxicity remains a critical issue in clinical oncology. We hypothesized that flavaglines, a family of natural compounds that display potent neuroprotective effects, may also alleviate doxorubicin-induced cardiotoxicity. METHODOLOGY/PRINCIPAL FINDINGS: Our in vitro data established that a pretreatment with flavaglines significantly increased viability of doxorubicin-injured H9c2 cardiomyocytes as demonstrated by annexin V, TUNEL and active caspase-3 assays. We demonstrated also that phosphorylation of the small heat shock protein Hsp27 is involved in the mechanism by which flavaglines display their cardioprotective effect. Furthermore, knocking-down Hsp27 in H9c2 cardiomyocytes completely reversed this cardioprotection. Administration of our lead compound (FL3) to mice attenuated cardiomyocyte apoptosis and cardiac fibrosis, as reflected by a 50% decrease of mortality. CONCLUSIONS/SIGNIFICANCE: These results suggest a prophylactic potential of flavaglines to prevent doxorubicin-induced cardiac toxicity.


Assuntos
Benzofuranos/farmacologia , Doxorrubicina/efeitos adversos , Proteínas de Choque Térmico HSP27/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Animais , Apoptose/efeitos dos fármacos , Benzofuranos/química , Cardiotônicos/química , Cardiotônicos/farmacologia , Meios de Cultura Livres de Soro , Citoproteção/efeitos dos fármacos , Fibrose , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação/efeitos dos fármacos
6.
J Med Chem ; 52(16): 5176-87, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19655762

RESUMO

Flavaglines constitute a family of natural anticancer compounds. We present here 3 (FL3), the first synthetic flavagline that inhibits cell proliferation and viability (IC(50) approximately 1 nM) at lower doses than did the parent compound, racemic rocaglaol. Compound 3 enhanced doxorubicin cytotoxicity in HepG2 cells and retained its potency against adriamycin-resistant cell lines without inducing cardiomyocyte toxicity. Compound 3 induced apoptosis of HL60 and Hela cells by triggering the translocation of Apoptosis Inducing Factor (AIF) and caspase-12 to the nucleus. A fluorescent conjugate of 3 accumulated in endoplasmic reticulum (ER), suggesting that flavaglines bind to their target in the ER, where it triggers a cascade of events that leads to the translocation of AIF and caspase-12 to the nucleus and probably inhibition of eIF4A. Our studies highlight structural features critical to their antineoplastic potential and suggest that these compounds would retain their activity in cells refractory to caspase activation.


Assuntos
Antineoplásicos/síntese química , Fator de Indução de Apoptose/metabolismo , Benzofuranos/síntese química , Caspase 12/metabolismo , Transporte Ativo do Núcleo Celular , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose , Benzofuranos/química , Benzofuranos/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Retículo Endoplasmático/metabolismo , Fase G2/efeitos dos fármacos , Humanos , Estereoisomerismo , Relação Estrutura-Atividade
7.
Mol Ther ; 12(5): 803-12, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16112910

RESUMO

In primary glioblastomas and other tumor types, the epidermal growth factor receptor (EGFR) is frequently observed with alterations, such as amplification, structural rearrangements, or overexpression of the gene, suggesting an important role in glial tumorigenesis and progression. In this study, we investigated whether posttranscriptional gene silencing by vector-mediated RNAi to inhibit EGFR expression can reduce the growth of cultured human gli36 glioma cells. To "knock down" EGFR expression, we have created HSV-1-based amplicons that contain the RNA polymerase III-dependent H1 promoter to express double-stranded hairpin RNA directed against EGFR at two different locations (pHSVsiEGFR I and pHSVsiEGFR II). We demonstrate that both pHSVsiEGFR I and pHSVsiEGFR II mediated knock-down of transiently transfected full-length EGFR or endogenous EGFR in a dose-dependent manner. The knock-down of EGFR resulted in the growth inhibition of human glioblastoma (gli36-luc) cells both in culture and in athymic mice in vivo. Cell cycle analysis and annexin V staining revealed that siRNA-mediated suppression of EGFR induced apoptosis. Overall HSV-1 amplicons can mediate efficient and specific posttranscriptional gene silencing.


Assuntos
Receptores ErbB/antagonistas & inibidores , Terapia Genética , Glioblastoma/terapia , Herpesvirus Humano 1/genética , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Transferência de Genes , Genes erbB-1 , Vetores Genéticos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA