Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 29(4): 1208-19, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25466901

RESUMO

We have previously shown that TNF-tumor necrosis factor receptor-2/p75 (TNFR2/p75) signaling plays a critical role in ischemia-induced neovascularization in skeletal muscle and heart tissues. To determine the role of TNF-TNFR2/p75 signaling in ischemia-induced inflammation and muscle regeneration, we subjected wild-type (WT) and TNFR2/p75 knockout (p75KO) mice to hind limb ischemia (HLI) surgery. Ischemia induced significant and long-lasting inflammation associated with considerable decrease in satellite-cell activation in p75KO muscle tissue up to 10 d after HLI surgery. To determine the possible additive negative roles of tissue aging and the absence of TNFR2/p75, either in the tissue or in the bone marrow (BM), we generated 2 chimeric BM transplantation (BMT) models where both young green fluorescent protein (GFP)-positive p75KO and WT BM-derived cells were transplanted into adult p75KO mice. HLI surgery was performed 1 mo after BMT, after confirming complete engraftment of the recipient BM with GFP donor cells. In adult p75KO with the WT-BMT, proliferative (Ki67(+)) cells were detected only by d 28 and were exclusively GFP(+), suggesting significantly delayed contribution of young WT-BM cell to adult p75KO ischemic tissue recovery. No GFP(+) young p75KO BM cells survived in adult p75KO tissue, signifying the additive negative roles of tissue aging combined with decreased/absent TNFR2/p75 signaling in postischemic recovery.


Assuntos
Isquemia/patologia , Isquemia/fisiopatologia , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Receptores Tipo II do Fator de Necrose Tumoral/genética , Células Satélites de Músculo Esquelético/patologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Apoptose , Transplante de Medula Óssea , Proliferação de Células , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Membro Posterior/irrigação sanguínea , Inflamação/etiologia , Inflamação/patologia , Inflamação/fisiopatologia , Isquemia/terapia , Macrófagos/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neovascularização Fisiológica , Infiltração de Neutrófilos , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes/genética , Regeneração , Transdução de Sinais
2.
J Biol Chem ; 289(23): 16200-13, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24742672

RESUMO

Adiponectin is a well described anti-inflammatory adipokine that is highly abundant in serum. Previous reports have found that adiponectin deficiency promotes cardiovascular and metabolic dysfunction in murine models, whereas its overexpression is protective. Two candidate adiponectin receptors, AdipoR1 and AdipoR2, are uncharacterized with regard to cardiovascular tissue homeostasis, and their in vivo metabolic functions remain controversial. Here we subjected AdipoR1- and AdipoR2-deficient mice to chronic hind limb ischemic surgery. Blood flow recovery in AdipoR1-deficient mice was similar to wild-type; however, revascularization in AdipoR2-deficient mice was severely attenuated. Treatment with adiponectin enhanced the recovery of wild-type mice but failed to rescue the impairment observed in AdipoR2-deficient mice. In view of this divergent receptor function in the hind limb ischemia model, AdipoR1- and AdipoR2-deficient mice were also evaluated in a model of diet-induced obesity. Strikingly, AdipoR1-deficient mice developed severe metabolic dysfunction compared with wild type, whereas AdipoR2-deficient mice were protected from diet-induced weight gain and metabolic perturbations. These data show that AdipoR2, but not AdipoR1, is functionally important in an in vivo model of ischemia-induced revascularization and that its expression is essential for the revascularization actions of adiponectin. These data also show that, in contrast to revascularization responses, AdipoR1, but not AdipoR2 deficiency, leads to diet-induced metabolic dysfunction, revealing that these receptors have highly divergent roles in vascular and metabolic homeostasis.


Assuntos
Doenças Metabólicas/fisiopatologia , Neovascularização Fisiológica , Receptores de Adiponectina/fisiologia , Animais , Extremidades/irrigação sanguínea , Camundongos , Camundongos Knockout
3.
J Biol Chem ; 289(29): 19907-16, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24920669

RESUMO

The sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA) is key to Ca(2+) homeostasis and is redox-regulated by reversible glutathione (GSH) adducts on the cysteine (C) 674 thiol that stimulate Ca(2+) uptake activity and endothelial cell angiogenic responses in vitro. We found that mouse hind limb muscle ischemia induced S-glutathione adducts on SERCA in both whole muscle tissue and endothelial cells. To determine the role of S-glutathiolation, we used a SERCA 2 C674S heterozygote knock-in (SKI) mouse lacking half the key thiol. Following hind limb ischemia, SKI animals had decreased SERCA S-glutathione adducts and impaired blood flow recovery. We studied SKI microvascular endothelial cells in which total SERCA 2 expression was unchanged. Cultured SKI microvascular endothelial cells showed impaired migration and network formation compared with wild type (WT). Ca(2+) studies showed decreased nitric oxide (·NO)-induced (45)Ca(2+) uptake into the endoplasmic reticulum (ER) of SKI cells, while Fura-2 studies revealed lower Ca(2+) stores and decreased vascular endothelial growth factor (VEGF)- and ·NO-induced Ca(2+) influx. Adenoviral overexpression of calreticulin, an ER Ca(2+) binding protein, increased ionomycin-releasable stores, VEGF-induced Ca(2+) influx and endothelial cell migration. Taken together, these data indicate that the redox-sensitive Cys-674 thiol on SERCA 2 is required for normal endothelial cell Ca(2+) homeostasis and ischemia-induced angiogenic responses, revealing a novel redox control of angiogenesis via Ca(2+) stores.


Assuntos
Cálcio/metabolismo , Glutationa/análogos & derivados , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Animais , Sinalização do Cálcio , Células Endoteliais/metabolismo , Feminino , Técnicas de Introdução de Genes , Glutationa/metabolismo , Hemodinâmica , Membro Posterior/irrigação sanguínea , Hipóxia/enzimologia , Hipóxia/fisiopatologia , Isquemia/enzimologia , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/enzimologia , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neovascularização Fisiológica , Óxido Nítrico/metabolismo , Oxirredução , Gravidez , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
J Biol Chem ; 288(34): 24886-97, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23824191

RESUMO

Adipose tissue secretes protein factors that have systemic actions on cardiovascular tissues. Previous studies have shown that ablation of the adipocyte-secreted protein adiponectin leads to endothelial dysfunction, whereas its overexpression promotes wound healing. However, the receptor(s) mediating the protective effects of adiponectin on the vasculature is not known. Here we examined the role of membrane protein T-cadherin, which localizes adiponectin to the vascular endothelium, in the revascularization response to chronic ischemia. T-cadherin-deficient mice were analyzed in a model of hind limb ischemia where blood flow is surgically disrupted in one limb and recovery is monitored over 28 days by laser Doppler perfusion imaging. In this model, T-cadherin-deficient mice phenocopy adiponectin-deficient mice such that both strains display an impaired blood flow recovery compared with wild-type controls. Delivery of exogenous adiponectin rescued the impaired revascularization phenotype in adiponectin-deficient mice but not in T-cadherin-deficient mice. In cultured endothelial cells, T-cadherin deficiency by siRNA knockdown prevented the ability of adiponectin to promote cellular migration and proliferation. These data highlight a previously unrecognized role for T-cadherin in limb revascularization and show that it is essential for mediating the vascular actions of adiponectin.


Assuntos
Adiponectina/metabolismo , Caderinas/metabolismo , Endotélio Vascular/metabolismo , Neovascularização Fisiológica/fisiologia , Adiponectina/genética , Animais , Caderinas/genética , Técnicas de Silenciamento de Genes , Membro Posterior/irrigação sanguínea , Isquemia/genética , Isquemia/metabolismo , Camundongos , Camundongos Knockout
5.
Arterioscler Thromb Vasc Biol ; 33(12): 2867-76, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24072696

RESUMO

OBJECTIVE: Dystrophin, the missing or defective protein in Duchenne muscular dystrophy, is expressed not only in muscle cells but also in vascular endothelial cells (ECs). In this study, we assessed the effects of dystrophin deficiency on the angiogenic capacities of ECs. APPROACH AND RESULTS: We isolated vascular ECs from mdx mice, the murine equivalent of Duchenne muscular dystrophy in humans, and wild-type controls, and we found that mdx-derived ECs have impaired angiogenic properties, in terms of migration, proliferation, and tube formation. They also undergo increased apoptosis in vitro compared with wild-type cells and have increased senescence-associated ß-galactosidase activity. Mdx-derived ECs also display reduced ability to support myoblast proliferation when cocultured with satellite cell-derived primary myoblasts. These endothelial defects are mirrored by systemic impairment of angiogenesis in vivo, both on induction of ischemia, stimulation with growth factors in the corneal model and matrigel plug assays, and tumor growth. We also found that dystrophin forms a complex with endothelial NO synthase and caveolin-1 in ECs, and that NO production and cGMP formation are compromised in ECs isolated from mdx mice. Interestingly, treatment with aspirin enhances production of both cGMP and NO in dystrophic ECs, whereas low-dose aspirin improves the dystrophic phenotype of mdx mice in vivo, in terms of resistance to physical exercise, muscle fiber permeability, and capillary density. CONCLUSIONS: These findings demonstrate that impaired angiogenesis is a novel player and potential therapeutic target in Duchenne muscular dystrophy.


Assuntos
Distrofina/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Neovascularização Fisiológica , Animais , Apoptose , Aspirina/farmacologia , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Caveolina 1/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Senescência Celular , Técnicas de Cocultura , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Neovascularização da Córnea/fisiopatologia , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Distrofina/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Isquemia/metabolismo , Isquemia/patologia , Isquemia/fisiopatologia , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Mutação , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Neovascularização Patológica , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fatores de Tempo
6.
J Exp Med ; 203(1): 153-63, 2006 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-16401693

RESUMO

The cell surface receptor alpha4 integrin plays a critical role in the homing, engraftment, and maintenance of hematopoietic progenitor cells (HPCs) in the bone marrow (BM). Down-regulation or functional blockade of alpha4 integrin or its ligand vascular cell adhesion molecule-1 mobilizes long-term HPCs. We investigated the role of alpha4 integrin in the mobilization and homing of BM endothelial progenitor cells (EPCs). EPCs with endothelial colony-forming activity in the BM are exclusively alpha4 integrin-expressing cells. In vivo, a single dose of anti-alpha4 integrin antibody resulted in increased circulating EPC counts for 3 d. In hindlimb ischemia and myocardial infarction, systemically administered anti-alpha4 integrin antibody increased recruitment and incorporation of BM EPCs in newly formed vasculature and improved functional blood flow recovery and tissue preservation. Interestingly, BM EPCs that had been preblocked with anti-alpha4 integrin ex vivo or collected from alpha4 integrin-deficient mice incorporated as well as control cells into the neovasculature in ischemic sites, suggesting that alpha4 integrin may be dispensable or play a redundant role in EPC homing to ischemic tissue. These data indicate that functional disruption of alpha4 integrin may represent a potential angiogenic therapy for ischemic disease by increasing the available circulating supply of EPCs.


Assuntos
Integrina alfa4/metabolismo , Isquemia Miocárdica/fisiopatologia , Neovascularização Fisiológica , Células-Tronco/fisiologia , Animais , Medula Óssea , Movimento Celular , Células Endoteliais , Integrina alfa4/genética , Masculino , Camundongos , Camundongos Knockout
7.
Nat Med ; 11(11): 1197-204, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16244652

RESUMO

Sonic hedgehog (Shh) is a crucial regulator of organ development during embryogenesis. We investigated whether intramyocardial gene transfer of naked DNA encoding human Shh (phShh) could promote a favorable effect on recovery from acute and chronic myocardial ischemia in adult animals, not only by promoting neovascularization, but by broader effects, consistent with the role of this morphogen in embryogenesis. After Shh gene transfer, the hedgehog pathway was upregulated in mammalian fibroblasts and cardiomyocytes. This resulted in preservation of left ventricular function in both acute and chronic myocardial ischemia by enhanced neovascularization, and reduced fibrosis and cardiac apoptosis. Shh gene transfer also enhanced the contribution of bone marrow-derived endothelial progenitor cells to myocardial neovascularization. These data suggest that Shh gene therapy may have considerable therapeutic potential in individuals with acute and chronic myocardial ischemia by triggering expression of multiple trophic factors and engendering tissue repair in the adult heart.


Assuntos
Terapia Genética , Coração/embriologia , Miocárdio/metabolismo , Transdução de Sinais , Transativadores/uso terapêutico , Doença Aguda , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Doença Crônica , Modelos Animais de Doenças , Ecocardiografia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog , Humanos , Camundongos , Camundongos Mutantes , Isquemia Miocárdica/etiologia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Função Ventricular Esquerda/fisiologia
8.
Mol Ther ; 19(4): 658-66, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21224834

RESUMO

We have previously shown that the signaling pathway of the embryonic morphogen Sonic hedgehog (Shh) is recapitulated in the postnatal skeletal muscle in response to ischemia. We have also demonstrated that Shh is an indirect angiogenic agent upregulating various families of angiogenic growth factors and that Shh gene therapy improves angiogenesis and heart function in experimental models of myocardial ischemia. Based on these findings, we hypothesized that Shh gene therapy is beneficial in an experimental model of peripheral ischemia. We found that intramuscular (i.m.) treatment with a plasmid encoding the Shh human gene (phShh) increased blood flow, capillary density, and arteriole density in mice in which peripheral circulation of the hindlimb was disrupted by removal of the common femoral artery. Shh gene therapy also enhanced vasculogenesis, by increasing the number of circulating bone marrow (BM)-derived endothelial precursors and improving the contribution of these cells to the process of neovascularization. Finally, phShh treatment induced upregulation of prototypical angiogenic, arteriogenic, and vasculogenic factors, such as vascular endothelial growth factor (VEGF), angiopoietin 1 (Ang-1), and stromal cell-derived factor-1 (SDF-1α). These data suggest that Shh gene therapy merits further investigation for its ability to trigger the expression of potent trophic factors and stimulate pleiotropic aspects of neovascularization in the setting of ischemia.


Assuntos
Terapia Genética/métodos , Proteínas Hedgehog/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Angiopoietina-1/metabolismo , Animais , Quimiocina CXCL12/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteínas Hedgehog/genética , Isquemia/genética , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Circulation ; 119(5): 699-708, 2009 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19171856

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) are known to promote neovascularization in ischemic diseases. Recent evidence suggested that diabetic neuropathy is causally related to impaired angiogenesis and deficient growth factors. Accordingly, we investigated whether diabetic neuropathy could be reversed by local transplantation of EPCs. METHODS AND RESULTS: We found that motor and sensory nerve conduction velocities, blood flow, and capillary density were reduced in sciatic nerves of streptozotocin-induced diabetic mice but recovered to normal levels after hind-limb injection of bone marrow-derived EPCs. Injected EPCs were preferentially and durably engrafted in the sciatic nerves. A portion of engrafted EPCs were uniquely localized in close proximity to vasa nervorum, and a smaller portion of these EPCs were colocalized with endothelial cells. Multiple angiogenic and neurotrophic factors were significantly increased in the EPC-injected nerves. These dual angiogenic and neurotrophic effects of EPCs were confirmed by higher proliferation of Schwann cells and endothelial cells cultured in EPC-conditioned media. CONCLUSIONS: We demonstrate for the first time that bone marrow-derived EPCs could reverse various manifestations of diabetic neuropathy. These therapeutic effects were mediated by direct augmentation of neovascularization in peripheral nerves through long-term and preferential engraftment of EPCs in nerves and particularly vasa nervorum and their paracrine effects. These findings suggest that EPC transplantation could represent an innovative therapeutic option for treating diabetic neuropathy.


Assuntos
Diabetes Mellitus Experimental/complicações , Neuropatias Diabéticas/terapia , Células Endoteliais/citologia , Transplante de Células-Tronco Hematopoéticas , Nervo Isquiático/irrigação sanguínea , Animais , Apoptose , Células da Medula Óssea/citologia , Divisão Celular , Células Cultivadas , Neuropatias Diabéticas/fisiopatologia , Células-Tronco Hematopoéticas/citologia , Fluxometria por Laser-Doppler , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Condução Nervosa , Células de Schwann/citologia , Nervo Isquiático/fisiologia
10.
Am J Physiol Heart Circ Physiol ; 297(6): H1974-83, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19801490

RESUMO

Neuregulin-1 (NRG1) is a potential therapeutic agent for the treatment of doxorubicin (Dox)-induced heart failure. NRG1, however, activates the erbB2 receptor, which is frequently overexpressed in breast cancers. It is, therefore, important to understand how NRG1, via erbB2, protects the heart against Dox cardiotoxicity. Here, we studied NRG1-erbB2 signaling in Dox-treated mice hearts and in isolated neonatal rat ventricular myocytes (NRVM). Male C57BL/6 mice were treated with recombinant NRG1 before and daily after a single dose of Dox. Cardiac function was determined by catheterization. Two-week survival was analyzed by the Kaplan-Meier method. Cardiac troponins [cardiac troponin I (cTnI) and cardiac troponin T (cTnT)] and phosphorylated Akt protein levels were determined in mice hearts and in NRVM by Western blot analysis. Activation of caspases and ubiquitinylation of troponins were determined in NRVM by caspase assay and immunoprecipitation. NRG1 significantly improved survival and cardiac function in Dox-treated mice. NRG1 reduced the decrease in cTnI, cTnT, and cardiac troponin C (cTnC) and maintained Akt phosphorylation in Dox-treated mice hearts. NRG1 reduced the decrease in cTnI and cTnT mRNA and proteins in Dox-treated NRVM. Inhibition of erbB2, phosphoinositide 3-kinase (PI3K), Akt, and mTOR blocked the protective effects of NRG1 on cTnI and cTnT in NRVM. NRG1 significantly reduced Dox-induced caspase activation, which degraded troponins, in NRVM. NRG1 reduced Dox-induced proteasome degradation of cTnI. NRG1 attenuates Dox-induced decrease in cardiac troponins by increasing transcription and translation and by inhibiting caspase activation and proteasome degradation of troponin proteins. NRG1 maintains cardiac troponins by the erbB2-PI3K pathway, which may lessen Dox-induced cardiac dysfunction.


Assuntos
Fármacos Cardiovasculares/administração & dosagem , Cardiopatias/prevenção & controle , Miocárdio/metabolismo , Neuregulina-1/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Troponina I/metabolismo , Troponina T/metabolismo , Animais , Animais Recém-Nascidos , Antibióticos Antineoplásicos , Biomarcadores/sangue , Caspases/metabolismo , Células Cultivadas , Creatina Quinase/sangue , Modelos Animais de Doenças , Doxorrubicina , Glicoproteínas/metabolismo , Cardiopatias/induzido quimicamente , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Humanos , Injeções Subcutâneas , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptor ErbB-2 , Proteínas Recombinantes/administração & dosagem , Serina-Treonina Quinases TOR , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Troponina I/sangue , Troponina I/genética , Troponina T/sangue , Troponina T/genética , Ubiquitinação , Regulação para Cima , Função Ventricular Esquerda/efeitos dos fármacos
11.
Circ Res ; 100(1): 70-8, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17158336

RESUMO

Notch receptors are important mediators of cell fate during embryogenesis, but their role in adult physiology, particularly in postnatal angiogenesis, remains unknown. Of the Notch receptors, only Notch1 and Notch4 are expressed in vascular endothelial cells. Here we show that blood flow recovery and postnatal neovascularization in response to hindlimb ischemia in haploinsufficient global or endothelial-specific Notch1(+/-) mice, but not Notch4(-/-) mice, were impaired compared with wild-type mice. The expression of vascular endothelial growth factor (VEGF) in response to ischemia was comparable between wild-type and Notch mutant mice, suggesting that Notch1 is downstream of VEGF signaling. Treatment of endothelial cells with VEGF increases presenilin proteolytic processing, gamma-secretase activity, Notch1 cleavage, and Hes-1 (hairy enhancer of split homolog-1) expression, all of which were blocked by treating endothelial cells with inhibitors of phosphatidylinositol 3-kinase/protein kinase Akt or infecting endothelial cells with a dominant-negative Akt mutant. Indeed, inhibition of gamma-secretase activity leads to decreased angiogenesis and inhibits VEGF-induced endothelial cell proliferation, migration, and survival. Overexpression of the active Notch1 intercellular domain rescued the inhibitory effects of gamma-secretase inhibitors on VEGF-induced angiogenesis. These findings indicate that the phosphatidylinositol 3-kinase/Akt pathway mediates gamma-secretase and Notch1 activation by VEGF and that Notch1 is critical for VEGF-induced postnatal angiogenesis. These results suggest that Notch1 may be a novel therapeutic target for improving angiogenic response and blood flow recovery in ischemic limbs.


Assuntos
Animais Recém-Nascidos , Endotélio Vascular/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Neovascularização Fisiológica , Receptor Notch1/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Bovinos , Movimento Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ativação Enzimática , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Notch1/química , Receptor Notch1/efeitos dos fármacos , Fluxo Sanguíneo Regional , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/farmacologia
12.
Circulation ; 115(6): 752-62, 2007 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-17261656

RESUMO

BACKGROUND: Aging is a risk factor for coronary and peripheral artery disease. Tumor necrosis factor-alpha (TNF-alpha), a proinflammatory cytokine, is expressed in ischemic tissue and is known to modulate angiogenesis. Little is known about the role of TNF-alpha receptors (TNFR1/p55 and TNFR2/p75) in angiogenic signaling. METHODS AND RESULTS: We studied neovascularization in the hindlimb ischemia model in young and old TNFR2/p75 knockout (p75KO) and wild-type age-matched controls. Between days 7 to 10 after hindlimb surgery, 100% of old p75KOs experienced autoamputation of the operated limbs, whereas none of the age-matched wild-type mice exhibited hindlimb necrosis. Poor blood flow recovery in p75KO mice was associated with increased endothelial cell apoptosis, decreased capillary density, and significant reductions in the expression of vascular endothelial growth factor and basic fibroblast growth factor-2 mRNA transcripts in ischemic tissue and in circulating endothelial progenitor cells. The number of circulating bone marrow-derived endothelial progenitor cells was significantly reduced in p75KO mice. Transplantation of wild-type bone marrow mononuclear cells into irradiated old p75KO mice 1 month before hindlimb surgery prevented limb loss. CONCLUSIONS: Our present study suggests that ischemia-induced endothelial progenitor cell-mediated neovascularization is dependent, at least in part, on p75 TNF receptor expressed in bone marrow-derived cells. Specifically, endothelial cell/endothelial progenitor cell survival, vascular endothelial growth factor expression, endothelial progenitor cell mobilization from bone marrow, endothelial progenitor cell differentiation, and ultimately ischemia-induced collateral vessel development are dependent on signaling through TNFR2/p75. Furthermore, because TNFR2/p75 becomes an age-related limiting factor in postischemic recovery, it may be a potential gene target for therapeutic interventions in adult vascular diseases.


Assuntos
Isquemia/fisiopatologia , Neovascularização Fisiológica , Receptores Tipo II do Fator de Necrose Tumoral/fisiologia , Envelhecimento/fisiologia , Animais , Apoptose , Transplante de Medula Óssea , Células Cultivadas , Células Endoteliais/patologia , Membro Posterior/irrigação sanguínea , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Transdução de Sinais , Células-Tronco/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética
13.
J Clin Invest ; 115(7): 1785-96, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15965500

RESUMO

TNF-alpha modulates EC proliferation and thereby plays a central role in new blood vessel formation in physiologic and pathologic circumstances. TNF-alpha is known to downregulate cyclin A, a key cell cycle regulatory protein, but little else is known about how TNF-alpha modulates EC cell cycle and angiogenesis. Using primary ECs, we show that ezrin, previously considered to act primarily as a cytoskeletal protein and in cytoplasmic signaling, is a TNF-alpha-induced transcriptional repressor. TNF-alpha exposure leads to Rho kinase-mediated phosphorylation of ezrin, which translocates to the nucleus and binds to cell cycle homology region repressor elements within the cyclin A promoter. Overexpression of dominant-negative ezrin blocks TNF-alpha-induced modulation of ezrin function and rescues cyclin A expression and EC proliferation. In vivo, blockade of ezrin leads to enhanced transplanted EC proliferation and angiogenesis in a mouse hind limb ischemia model. These observations suggest that TNF-alpha regulates angiogenesis via Rho kinase induction of a transcriptional repressor function of the cytoskeletal protein ezrin and that ezrin may represent a suitable therapeutic target for processes dependent on EC proliferation.


Assuntos
Ciclina A/genética , Proteínas do Citoesqueleto/fisiologia , Células Endoteliais/citologia , Fosfoproteínas/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Bovinos , Proliferação de Células , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Células Endoteliais/fisiologia , Células Endoteliais/transplante , Extremidades , Expressão Gênica , Humanos , Isquemia/terapia , Camundongos , Camundongos Nus , Neovascularização Fisiológica , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Transcrição Gênica , Transfecção
14.
J Vasc Res ; 45(5): 437-44, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18418002

RESUMO

The vascular endothelial growth factor/vascular endothelial growth factor receptor 2 (VEGF/VEGFR-2) signal transduction system plays a key role during embryonic vascular development and adult neovascularization. In contrast to many endothelial genes, VEGFR-2 is expressed at low levels in most adult vessels but is strongly upregulated during neovascularization, leading to a pro-angiogenic response. Here, we analyzed the activity of regulatory sequences of the murine Vegfr2 gene during neovessel formation in vivo under ischemic and inflammatory conditions. Hindlimb ischemia was induced in transgenic mice, expressing the LacZ reporter gene under the control of Vegfr2 promoter/enhancer elements. Most vessels in the ischemic muscle tissue showed strong endothelium-specific reporter gene expression, whereas nearly no LacZ-expressing capillaries were observed in untreated control tissue. Cutaneous punch wounds were created to induce angiogenesis under inflammatory conditions, leading to robust LacZ expression in the majority of the blood vessels in the wound tissue. Since the cornea is physiologically avascular, the functionality of these promoter/enhancer elements exclusively in newly formed vessels was confirmed using the cornea micropocket assay. Taken together, our results show that these Vegfr2 regulatory elements are active during adult neovessel formation in general. Therefore, these sequences may prove to be valuable targets for novel endothelium-specific anti-angiogenic as well as pro-angiogenic treatment strategies. They may especially allow directing therapeutic gene expression to sites of adult neovascularization. Moreover, the Vegfr2/LacZ reporter mice represent a powerful model to generally analyze the transcriptional control mechanisms involved in the induction of Vegfr2 expression during adult neovascularization.


Assuntos
Córnea/irrigação sanguínea , Neovascularização da Córnea/fisiopatologia , Genes Reporter , Inflamação/fisiopatologia , Isquemia/fisiopatologia , Óperon Lac , Músculo Esquelético/irrigação sanguínea , Neovascularização Patológica/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Neovascularização da Córnea/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Elementos Facilitadores Genéticos , Inflamação/metabolismo , Isquemia/metabolismo , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/metabolismo , Regiões Promotoras Genéticas , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Cicatrização
15.
Circ Res ; 98(5): 697-704, 2006 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-16484619

RESUMO

Delayed reendothelialization contributes to restenosis after angioplasty and stenting in diabetes. Prior data have shown that bone marrow (BM)-derived endothelial progenitor cells (EPCs) contribute to endothelial recovery after arterial injury. We investigated the hypothesis that the EPC contribution to reendothelialization may be impaired in diabetes, resulting in delayed reendothelialization. Reendothelialization was significantly reduced in diabetic mice compared with nondiabetic mice in a wire-induced carotid denudation model. The EPC contribution to neoendothelium was significantly reduced in Tie2/LacZ BM-transplanted diabetic versus nondiabetic mice. BM from diabetic and nondiabetic mice was transplanted into nondiabetic mice, revealing that reendothelialization was impaired in the recipients of diabetic BM. To examine the relative roles of denuded artery versus EPCs in diabetes, we injected diabetic and nondiabetic EPCs intravenously after arterial injury in diabetic and nondiabetic mice. Diabetic EPCs recruitment to the neoendothelium was significantly reduced, regardless of the diabetic status of the recipient mice. In vitro, diabetic EPCs exhibited decreased migration and adhesion activities. Vascular endothelial growth factor and endothelial NO synthase expressions were also significantly reduced in diabetic EPCs. Notably, thrombospondin-1 mRNA expression was significantly upregulated in diabetic EPCs, associating with the decreased EPC adhesion activity in vitro and in vivo. Reendothelialization is impaired by malfunctioning EPCs in diabetes. Diabetic EPCs have phenotypic differences involving thrombospondin-1 expression compared with nondiabetic EPCs, revealing potential novel mechanistic insights and therapeutic targets to improve reendothelialization and reduce restenosis in diabetes.


Assuntos
Diabetes Mellitus/fisiopatologia , Células Endoteliais/fisiologia , Células-Tronco/fisiologia , Trombospondina 1/fisiologia , Animais , Transplante de Medula Óssea , Adesão Celular , Movimento Celular , Células Cultivadas , Citocinas/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
16.
Mol Ther ; 15(1): 69-75, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17164777

RESUMO

Toxic neuropathy represents an important clinical problem in the use of the chemotherapeutic substances Taxol and thalidomide. Sensory neuropathy has a high incidence, lacks an effective treatment and is the dose-limiting factor for these drugs. The pathogenic basis of these neuropathies is unknown. We investigated the hypothesis that the experimental toxic neuropathies from Taxol and thalidomide results from destruction of vasa nervorum and can be reversed by the administration of an angiogenic cytokine. In animal models of Taxol- and thalidomide-induced neuropathy, nerve blood flow has been attenuated and the number of vasa nervorum has been reduced. Intramuscular gene transfer of naked plasmid DNA encoding VEGF-1 administered in parallel with Taxol injections completely inhibited deterioration of nerve function and diminution of the peripheral nerve vasculature. Gene therapy in animals with established Taxol- or thalidomide-induced neuropathies resulted in recovery of vascularity and improved nerve electrophysiology. These findings implicate microvascular damage as the basis for toxic neuropathy and suggest that angiogenic growth factors may constitute a novel treatment for this disorder.


Assuntos
Neovascularização Patológica/tratamento farmacológico , Paclitaxel/farmacologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Talidomida/farmacologia , Vasa Nervorum/efeitos dos fármacos , Vasa Nervorum/lesões , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Quimioterapia Combinada , Eletrofisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Humanos , Injeções Intramusculares , Lectinas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Plasmídeos/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Circulação Renal
17.
Circulation ; 114(20): 2163-9, 2006 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-17075009

RESUMO

BACKGROUND: We compared the therapeutic potential of purified mobilized human CD34+ cells with that of mobilized total mononuclear cells (tMNCs) for the preservation/recovery of myocardial tissue integrity and function after myocardial infarction (MI). METHODS AND RESULTS: CD34+ cells were purified from peripheral blood tMNCs of healthy volunteers by magnetic cell sorting after a 5-day administration of granulocyte colony-stimulating factor. Phosphate-buffered saline (PBS), 5x10(5) CD34+ cells/kg, 5x10(5) tMNCs/kg (low-dose MNCs [loMNCs]), or a higher dose of tMNCs (hiMNCs) containing 5x10(5) CD34+ cells/kg was transplanted intramyocardially 10 minutes after the induction of MI in athymic nude rats. Hematoxylin and eosin staining revealed that moderate to severe hemorrhagic MI on day 3 was more frequent in the hiMNC group than in the PBS and CD34+ cell groups. Immunostaining for human-specific CD45 revealed abundant distribution of hematopoietic/inflammatory cells derived from transplanted cells in the ischemic myocardium of the hiMNC group. Capillary density on day 28 was significantly greater in the CD34+ cell group (721.1+/-19.9 per 1 mm2) than in the PBS, loMNC, and hiMNC groups (384.7+/-11.0, 372.5+/-14.1, and 497.5+/-24.0 per 1 mm2) (P<0.01). Percent fibrosis area on day 28 was less in the CD34(+) cell group (15.6+/-0.9%) than in the PBS, loMNC, and hiMNC groups (26.3+/-1.2%, 27.5+/-1.8%, and 22.2+/-1.8%) (P<0.05). Echocardiographic fractional shortening on day 28 was significantly higher in the CD34+ cell group (30.3+/-0.9%) than in the PBS, loMNC, and hiMNC groups (22.7+/-1.5%, 23.4+/-1.1%, and 24.9+/-1.7%; P<0.05). Echocardiographic regional wall motion score was better preserved in the CD34+ cell group (21.8+/-0.5) than in the PBS, loMNC, and hiMNC groups (25.4+/-0.4, 24.9+/-0.4, and 24.1+/-0.6; P<0.05). CONCLUSIONS: CD34+ cells exhibit superior efficacy for preserving myocardial integrity and function after MI than unselected circulating MNCs.


Assuntos
Antígenos CD34/metabolismo , Monócitos/metabolismo , Monócitos/transplante , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Neovascularização Fisiológica , Animais , Diferenciação Celular , Ecocardiografia , Feminino , Hemorragia/etiologia , Humanos , Monócitos/patologia , Infarto do Miocárdio/complicações , Infarto do Miocárdio/patologia , Miocárdio/patologia , Período Pós-Operatório , Ratos , Ratos Nus , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Fatores de Tempo , Transplante Heterólogo , Função Ventricular Esquerda , Remodelação Ventricular
18.
Circulation ; 113(12): 1605-14, 2006 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-16534014

RESUMO

BACKGROUND: Recent data have indicated that estradiol can modulate the kinetics of endothelial progenitor cells (EPCs) via endothelial nitric oxide synthase (eNOS)-dependent mechanisms. We hypothesized that estradiol could augment the incorporation of bone marrow (BM)-derived EPCs into sites of ischemia-induced neovascularization, resulting in protection from ischemic injury. METHODS AND RESULTS: Myocardial infarction (MI) was induced by ligation of the left coronary artery in ovariectomized mice receiving either 17beta-estradiol or placebo. Estradiol induced significant increases in circulating EPCs 2 and 3 weeks after MI in estradiol-treated animals, and capillary density was significantly greater in estradiol-treated animals. Greater numbers of BM-derived EPCs were observed at ischemic sites in estradiol-treated animals than in placebo-treated animals 1 and 4 weeks after MI. In eNOS-null mice, the effect of estradiol on mobilization of EPCs was lost, as was the functional improvement in recovery from acute myocardial ischemia. A decrease was found in matrix metalloproteinase-9 (MMP-9) expression in eNOS-null mice under basal and estradiol-stimulated conditions after MI, the mobilization of EPCs by estradiol was lost in MMP-9-null mice, and the functional benefit conferred by estradiol treatment after MI in wild-type mice was significantly attenuated. CONCLUSIONS: Estradiol preserves the integrity of ischemic tissue by augmenting the mobilization and incorporation of BM-derived EPCs into sites of neovascularization by eNOS-mediated augmentation of MMP-9 expression in the BM. Moreover, these data have broader implications with regard to our understanding of the role of EPCs in post-MI recovery and on the sex discrepancy in cardiac events.


Assuntos
Células Endoteliais/fisiologia , Estradiol/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Infarto do Miocárdio/terapia , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Animais , Células da Medula Óssea/fisiologia , Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Mobilização de Células-Tronco Hematopoéticas/métodos , Metaloproteinase 9 da Matriz/fisiologia , Camundongos , Infarto do Miocárdio/tratamento farmacológico , Isquemia Miocárdica/patologia , Isquemia Miocárdica/prevenção & controle , Óxido Nítrico Sintase Tipo II/fisiologia , Óxido Nítrico Sintase Tipo III , Ovariectomia , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Resultado do Tratamento
19.
Circulation ; 114(21): 2261-70, 2006 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-17088460

RESUMO

BACKGROUND: Estradiol (E2) modulates the kinetics of circulating endothelial progenitor cells (EPCs) and favorably affects neovascularization after ischemic injury. However, the roles of estrogen receptors alpha (ER alpha) and beta (ER beta) in EPC biology are largely unknown. METHODS AND RESULTS: In response to E2, migration, tube formation, adhesion, and estrogen-responsive element-dependent gene transcription activities were severely impaired in EPCs obtained from ER alpha-knockout mice (ER alphaKO) and moderately impaired in ER betaKO EPCs. The number of ER alphaKO EPCs (42.4+/-1.5; P<0.001) and ER betaKO EPCs (55.4+/-1.8; P=0.03) incorporated into the ischemic border zone was reduced as compared with wild-type (WT) EPCs (72.5+/-1.3). In bone marrow transplantation (BMT) models, the number of mobilized endogenous EPCs in E2-treated mice was significantly reduced in ER alphaKO BMT (WT mice transplanted with ER alphaKO bone marrow) (2.03+/-0.18%; P=0.004 versus WT BMT) and ER betaKO BMT (2.62+/-0.07%; P=0.02 versus WT) compared with WT BMT (2.87+/-0.13%) (WT to WT BMT as control) mice. Capillary density at the border zone of ischemic myocardium also was significantly reduced in ER alphaKO BMT and ER betaKO BMT compared with WT mice (WT BMT, 1718+/-75/mm2; ER alphaKO BMT, 1107+/-48/mm2; ER betaKO BMT, 1567+/-50/mm2). ER alpha mRNA was expressed more abundantly on EPCs compared with ER beta. Moreover, vascular endothelial growth factor was significantly downregulated on ER alphaKO EPCs compared with WT EPCs both in vitro and in vivo. CONCLUSIONS: Both ER alpha and ER beta contribute to E2-mediated EPC activation and tissue incorporation and to preservation of cardiac function after myocardial infarction. ER alpha plays a more prominent role in this process. Moreover, ER alpha contributes to upregulation of vascular endothelial growth factor, revealing possible mechanisms of an effect of E2 on EPC biology. Finally, these data provide additional evidence of the importance of bone marrow-derived EPC phenotype in ischemic tissue repair.


Assuntos
Células da Medula Óssea/citologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Coração/fisiopatologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Transplante de Células-Tronco , Animais , Cardiotônicos/farmacologia , Diferenciação Celular , Células Cultivadas , Células Endoteliais/citologia , Estradiol/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/deficiência , Receptor beta de Estrogênio/deficiência , Feminino , Mobilização de Células-Tronco Hematopoéticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recuperação de Função Fisiológica , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Endocrinology ; 148(8): 3618-24, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17478555

RESUMO

Preclinical and observational studies in ovariectomized (OVX) animals and pre- and postmenopausal women, respectively, have suggested the cardioprotective effects of estrogen replacement therapy. However, randomized clinical trials have not confirmed estrogen-mediated cardioprotection. Although uncertainties about the duration and optimal type of estrogen replacement regimen might explain the disparity, other factors that may mask the protective effects of 17beta-estradiol (E2) on cardiovascular outcome need scrutiny. Increased ethanol consumption may be one such factor. We examined the effect of E2 supplementation on ethanol consumption in OVX mice and the effect of ethanol consumption on E2-mediated vascular repair, in vivo. OVX mice implanted with E2 pellets consumed significantly more ethanol, compared with those receiving placebo pellets. E2-induced increase in ethanol consumption was not affected by the absence of either estrogen receptor-alpha or -beta. Reendothelialization after carotid artery denudation was repressed, and neovascularization in ischemic hind limbs was blunted in mice consuming ethanol, despite E2 supplementation. In vitro, ethanol dose-dependently attenuated E2-induced endothelial cell (EC) proliferation and tube formation activity and enhanced EC apoptosis, suggesting that ethanol blocks E2-induced EC survival and function. Taken together our data suggest that increased ethanol consumption after E2 supplementation blunts the beneficial effects of E2 on EC function and that novel approaches to estrogen replacement for cardioprotection may benefit from the control of alcohol consumption.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Lesões das Artérias Carótidas/fisiopatologia , Estradiol/farmacologia , Terapia de Reposição de Estrogênios , Animais , Comportamento Animal/fisiologia , Lesões das Artérias Carótidas/cirurgia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Feminino , Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Ovariectomia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Volição/efeitos dos fármacos , Volição/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa