Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(3)2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-35163735

RESUMEN

Oncostatin M (OSM), a member of the interleukin-6 family, functions as a major mediator of cardiomyocyte remodeling under pathological conditions. Its involvement in a variety of human cardiac diseases such as aortic stenosis, myocardial infarction, myocarditis, cardiac sarcoidosis, and various cardiomyopathies make the OSM receptor (OSMR) signaling cascades a promising therapeutic target. However, the development of pharmacological treatment strategies is highly challenging for many reasons. In mouse models of heart disease, OSM elicits opposing effects via activation of the type II receptor complex (OSMR/gp130). Short-term activation of OSMR/gp130 protects the heart after acute injury, whereas chronic activation promotes the development of heart failure. Furthermore, OSM has the ability to integrate signals from unrelated receptors that enhance fetal remodeling (dedifferentiation) of adult cardiomyocytes. Because OSM strongly stimulates the production and secretion of extracellular proteins, it is likely to exert systemic effects, which in turn, could influence cardiac remodeling. Compared with the mouse, the complexity of OSM signaling is even greater in humans because this cytokine also activates the type I leukemia inhibitory factor receptor complex (LIFR/gp130). In this article, we provide an overview of OSM-induced cardiomyocyte remodeling and discuss the consequences of OSMR/gp130 and LIFR/gp130 activation under acute and chronic conditions.


Asunto(s)
Insuficiencia Cardíaca , Interleucina-6 , Miocitos Cardíacos , Oncostatina M , Receptores de Oncostatina M , Animales , Receptor gp130 de Citocinas/metabolismo , Humanos , Interleucina-6/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 40(5): e126-e137, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32188276

RESUMEN

OBJECTIVE: Arteriogenesis, describing the process of collateral artery growth, is activated by fluid shear stress (FSS). Since this vascular mechanotransduction may involve microRNAs (miRNAs), we investigated the FSS-induced expression of vascular cell miRNAs and their functional impact on collateral artery growth during arteriogenesis. Approach and Results: To this end, rats underwent femoral artery ligation and arteriovenous anastomosis to increase collateral blood flow to maximize FSS and trigger collateral vessel remodeling. Five days after surgery, a miRNA expression profile was obtained from collateral tissue, and upregulation of 4 miRNAs (miR-24-3p, miR-143-3p, miR-146a-5p, and miR-195-5p) was verified by quantitative polymerase chain reaction. Knockdown of miRNAs at the same time of the surgery in an in vivo mouse ligation and recovery model demonstrated that inhibition of miR-143-3p only severely impaired blood flow recovery due to decreased arteriogenesis. In situ hybridization revealed distinct localization of miR-143-3p in the vessel wall of growing collateral arteries predominantly in smooth muscle cells. To investigate the mechanotransduction of FSS leading to the increased miR-143-3p expression, cultured endothelial cells were exposed to FSS. This provoked the expression and release of TGF-ß (transforming growth factor-ß), which increased the expression of miR-143-3p in smooth muscle cells in the presence of SRF (serum response factor) and myocardin. COL5A2 (collagen type V-α2)-a target gene of miR-143-3p predicted by in silico analysis-was found to be downregulated in growing collaterals. CONCLUSIONS: These results indicate that the increased miR-143-3p expression in response to FSS might contribute to the reorganization of the extracellular matrix, which is important for vascular remodeling processes, by inhibiting collagen V-α2 biosynthesis.


Asunto(s)
Colágeno Tipo V/metabolismo , Circulación Colateral , Arteria Femoral/cirugía , Mecanotransducción Celular , MicroARNs/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Animales , Derivación Arteriovenosa Quirúrgica , Velocidad del Flujo Sanguíneo , Células Cultivadas , Colágeno Tipo V/genética , Arteria Femoral/metabolismo , Arteria Femoral/fisiopatología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ligadura , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Ratas Sprague-Dawley , Flujo Sanguíneo Regional , Estrés Mecánico
3.
Int J Mol Sci ; 21(17)2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32882982

RESUMEN

Fetal and hypertrophic remodeling are hallmarks of cardiac restructuring leading chronically to heart failure. Since the Ras/Raf/MEK/ERK cascade (MAPK) is involved in the development of heart failure, we hypothesized, first, that fetal remodeling is different from hypertrophy and, second, that remodeling of the MAPK occurs. To test our hypothesis, we analyzed models of cultured adult rat cardiomyocytes as well as investigated myocytes in the failing human myocardium by western blot and confocal microscopy. Fetal remodeling was induced through endothelial morphogens and monitored by the reexpression of Acta2, Actn1, and Actb. Serum-induced hypertrophy was determined by increased surface size and protein content of cardiomyocytes. Serum and morphogens caused reprogramming of Ras/Raf/MEK/ERK. In both models H-Ras, N-Ras, Rap2, B- and C-Raf, MEK1/2 as well as ERK1/2 increased while K-Ras was downregulated. Atrophy, MAPK-dependent ischemic resistance, loss of A-Raf, and reexpression of Rap1 and Erk3 highlighted fetal remodeling, while A-Raf accumulation marked hypertrophy. The knock-down of B-Raf by siRNA reduced MAPK activation and fetal reprogramming. In conclusion, we demonstrate that fetal and hypertrophic remodeling are independent processes and involve reprogramming of the MAPK.


Asunto(s)
Reprogramación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/citología , Remodelación Vascular , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/genética , Masculino , Proteínas Quinasas Activadas por Mitógenos/genética , Miocitos Cardíacos/metabolismo , Ratas , Ratas Wistar , Transducción de Señal
4.
Cells Tissues Organs ; 201(2): 118-29, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26796132

RESUMEN

We previously reported excessive growth of collateral vessels in the dog heart during arteriogenesis induced by implantation of an ameroid constrictor around the circumflex branch of the left coronary artery. In the present study, using histology and immunocofocal microscopy, we further investigated how these aberrant collateral vessels form. By comparison with mature collateral vessels the following findings were made: perivascular space was very narrow where damage of the perivascular myocardium occurred; the neointima was very thick, resulting in a very small lumen; elastica van Gieson staining revealed the absence of the internal elastic lamina and of elastic fibers in the adventitia, but abundant collagen in the adventitia as well as in the neointima; smooth muscle cells of the neointima expressed less α-SM actin and little desmin; expression of the fibroblast growth factors aFGF, bFGF and platelet-derived growth factor (PDGF)-AB was observed mainly in the endothelial cells and abluminal region, but transforming growth factor-ß1 was only present in the adventitia and damaged myocardium; angiogenesis in the neointima was observed in some collateral vessels expressing high levels of eNOS, and cell proliferation was mainly present in the abluminal region, but apoptosis was in the deep neointima. In conclusion, these data for the first time reveal that the formation of the aberrant collateral vessels in the dog heart involves active extracellular proteolysis and a special expression profile of growth factors, eNOS, cell proliferation and apoptosis. The finding of a narrow perivascular space and perivascular myocardial damage suggests that anatomical constraint is most likely the cause for exacerbated inward remodeling in aberrant collateral vessels in dog heart.


Asunto(s)
Oclusión Coronaria/fisiopatología , Vasos Coronarios/fisiopatología , Neovascularización Patológica/fisiopatología , Actinas/análisis , Animales , Apoptosis , Proliferación Celular , Oclusión Coronaria/patología , Vasos Coronarios/patología , Desmina/análisis , Perros , Péptidos y Proteínas de Señalización Intercelular/análisis , Miocardio/patología , Neovascularización Patológica/patología , Óxido Nítrico Sintasa de Tipo III/análisis
5.
Arterioscler Thromb Vasc Biol ; 35(3): 589-97, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25550202

RESUMEN

OBJECTIVE: Arteriogenesis is strongly dependent on the recruitment of leukocytes, especially monocytes, into the perivascular space of growing collateral vessels. On the basis of previous findings that platelets are central players in inflammatory processes and mediate the recruitment of leukocytes, the aim of this study was to assess the role of platelets in a model of arterial remodeling. APPROACH AND RESULTS: C57Bl6 wild-type mice, IL4-R/Iba mice lacking the extracellular domain of the glycoprotein Ibα (GPIbα) receptor, and mice treated with antibodies to block GPIbα or deplete circulating platelets were studied in peripheral arteriogenesis. Using a novel model of intravital 2-photon and epifluorescence imaging, we visualized and quantified the interaction of platelets with leukocytes and the vascular endothelium in vivo. We found that transient platelet adhesion to the endothelium of collateral vessels was a major event during arteriogenesis and depended on GPIbα. Furthermore, leukocyte recruitment was obviously affected in animals with defective platelet GPIbα function. In IL4-R/Iba mice, transient and firm leukocyte adhesion to the endothelium of collateral vessels, as well as leukocyte accumulation in the perivascular space, were significantly reduced. Furthermore, we detected platelet-leukocyte aggregates within the circulation, which were significantly reduced in IL4-R/Iba animals. Finally, platelet depletion and loss of GPIbα function resulted in poor reperfusion recovery as determined by laser Doppler imaging. CONCLUSIONS: Thus, GPIbα-mediated interactions between platelets and endothelial cells, as well as leukocytes, support innate immune cell recruitment and promote arteriogenesis-establishing platelets as critical players in this process.


Asunto(s)
Neovascularización Fisiológica , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Animales
6.
Diabetes Metab Res Rev ; 28 Suppl 1: 27-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22271719

RESUMEN

Different forms of vessel growth in the adult organism contribute to the compensation for an occluded artery. We here summarize the major differences between arteriogenesis and angiogenesis and provide evidence in favour of a therapeutic stimulation of collateral growth. In addition, we outline current knowledge about regulatory mechanisms transducing the initial physical stimulus into a cellular response. As an example, the role of nitric oxide during arteriogenesis is discussed, and finally, we propose a mechanism of how an efficient decision is made that makes the larger collaterals larger and the smaller ones smaller.


Asunto(s)
Arteriopatías Oclusivas/fisiopatología , Arteriopatías Oclusivas/terapia , Circulación Colateral , Neovascularización Fisiológica , Adulto , Humanos
7.
Circ Res ; 106(4): 695-704, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20044516

RESUMEN

RATIONALE: We previously discovered the human 10T-->C (Trp4Arg) missense mutation in exon 2 of the muscle LIM protein (MLP, CSRP3) gene. OBJECTIVE: We sought to study the effects of this single-nucleotide polymorphism in the in vivo situation. METHODS AND RESULTS: We now report the generation and detailed analysis of the corresponding Mlp(W4R/+) and Mlp(W4R/W4R) knock-in animals, which develop an age- and gene dosage-dependent hypertrophic cardiomyopathy and heart failure phenotype, characterized by almost complete loss of contractile reserve under catecholamine induced stress. In addition, evidence for skeletal muscle pathology, which might have implications for human mutation carriers, was observed. Importantly, we found significantly reduced MLP mRNA and MLP protein expression levels in hearts of heterozygous and homozygous W4R-MLP knock-in animals. We also detected a weaker in vitro interaction of telethonin with W4R-MLP than with wild-type MLP. These alterations may contribute to an increased nuclear localization of W4R-MLP, which was observed by immunohistochemistry. CONCLUSIONS: Given the well-known high frequency of this mutation in Caucasians of up to 1%, our data suggest that (W4R-MLP) might contribute significantly to human cardiovascular disease.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Insuficiencia Cardíaca/metabolismo , Proteínas Musculares/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Función Ventricular Izquierda , Factores de Edad , Envejecimiento , Animales , Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Células Cultivadas , Conectina , Modelos Animales de Enfermedad , Fibrosis , Técnicas de Sustitución del Gen , Genotipo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Heterocigoto , Homocigoto , Proteínas con Dominio LIM , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación Missense , Miocitos Cardíacos/patología , Fenotipo , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Transfección
8.
BMC Dev Biol ; 11: 28, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21595990

RESUMEN

BACKGROUND: The zinc finger transcription factor Egr-1 (Early growth response 1) is central to several growth factors and represents an important activator of target genes not only involved in physiological processes like embryogenesis and neonatal development, but also in a variety of pathophysiological processes, for example atherosclerosis or cancer. Current options to investigate its transcription and activation in vivo are end-point measurements that do not provide insights into dynamic changes in the living organism. RESULTS: We developed a transgenic mouse (Egr-1-luc) in which the luciferase reporter gene is under the control of the murine Egr-1 promoter providing a versatile tool to study the time course of Egr-1 activation in vivo. In neonatal mice, bioluminescence imaging revealed a high Egr-1 promoter activity reaching basal levels three weeks after birth with activity at snout, ears and paws. Using a model of partial hepatectomy we could show that Egr-1 promoter activity and Egr-1 mRNA levels were increased in the regenerating liver. In a model of wound healing, we demonstrated that Egr-1 promoter activity was upregulated at the site of injury. CONCLUSION: Taken together, we have developed a transgenic mouse model that allows real time in vivo imaging of the Egr-1 promoter activity. The ability to monitor and quantify Egr-1 activity in the living organism may facilitate a better understanding of Egr-1 function in vivo.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Embrión de Mamíferos/fisiología , Regeneración Hepática/fisiología , Regiones Promotoras Genéticas , Cicatrización de Heridas/fisiología , Animales , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Embrión de Mamíferos/anatomía & histología , Femenino , Genes Reporteros , Hepatectomía , Hígado/citología , Hígado/metabolismo , Hígado/patología , Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Mol Cell Biochem ; 354(1-2): 219-29, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21509579

RESUMEN

Innervation plays an important role in development and remodeling of blood vessels. However, very little is known whether innervation is involved in arteriogenesis. In the present study, we tested the hypothesis that innervation may contribute to the process of arteriogenesis induced by ligature of femoral artery in rat/rabbit hind limb with or without denervation. We found that: (1) angiography showed more collateral vessels in the ligature side than that in ligature plus denervation side; (2) collateral vessels in denervation side was characterized by an inward remodeling; (3) in both collateral vessels (CVs) from only femoral ligature side as well as the ligature plus denervation side, ICAM-1 and VCAM-1 expression was up-regulated but increased VCAM-1 was more evident in the adventitia of collateral vessels of only femoral ligature side; (4) 7 days after surgery, in CVs from the femoral ligature side only, numerous macrophages (RAM11 positive cells) and high cell proliferation ratio (ki67 positive cells) were detected, but they were less in the denervation side. In conclusion, our data demonstrate for the first time that neural regulation is one of the factors that contributes to collateral vessel growth in rat/rabbit hind limb ischemic model by showing collateral vessel growth induced by femoral artery ligature is impaired by denervation.


Asunto(s)
Circulación Colateral/fisiología , Arteria Femoral/cirugía , Miembro Posterior/irrigación sanguínea , Neovascularización Fisiológica , Nervio Ciático/cirugía , Angiografía , Animales , Proliferación Celular , Desnervación , Miembro Posterior/anatomía & histología , Miembro Posterior/inervación , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno Ki-67/metabolismo , Ligadura , Macrófagos/metabolismo , Proteínas de Neurofilamentos/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Mol Cell Biochem ; 343(1-2): 223-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20559689

RESUMEN

Growth factors are viewed as main arteriogenic stimulators for collateral vessel growth. However, the information about their native expression and distribution in collateral vessels is still limited. This study was designed to profile expression of acidic and basic FGF, platelet-derived growth factor (PDGF-AB) and vascular endothelial growth factor (VEGF-A) and its receptor, fetal liver kinase-1 (Flk-1) during arteriogenesis by confocal immunofluorescence in both dog ameroid constrictor model and rabbit arteriovenous shunt model of arteriogenesis. We found that: (1) in normal arteries (NA) in dog heart, aFGF, bFGF, and PDGF-AB all were mainly expressed in endothelial cells (EC) and media smooth muscle cells (SMC), but the expression of aFGF was very weak, with those of the other two being moderate; (2) in collateral arteries (CAs), aFGF, bFGF, and PDGF-AB all were significantly upregulated (P < 0.05); they were present in all the layers of the vascular wall and were 2.1, 1.7, and 1.9 times higher than that in NA, respectively; and (3) in NA in rabbit hind limb, VEGF-A was absent, Flk-1 was only weakly present in endothelial cells, but in one week CAs VEGF-A and Flk-1 were significantly increased in both shunt and ligation sides; this was more evident in the shunt-side CAs, 2.3, and 2 times higher than that in the ligation side, respectively. In conclusion, our data demonstrate for the first time that growth factors, aFGF, bFGF, and PDGF-AB are significantly upregulated in collateral vessels in dog heart, and enhanced VEGF-A and its receptor, Flk-1, are associated with rapid and lasting increased shear stress. These findings suggest that endogenous production of growth factors could be an important factor promoting collateral vessel growth.


Asunto(s)
Arterias/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Animales , Perros , Inmunohistoquímica , Conejos
11.
Arterioscler Thromb Vasc Biol ; 29(12): 2093-101, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19778941

RESUMEN

OBJECTIVE: Arteriogenesis, the development of a collateral circulation, is important for tissue survival but remains functionally defective because of early normalization of fluid shear stress (FSS). Using a surgical model of chronically elevated FSS we showed that rabbits exhibited normal blood flow reserve after femoral artery ligature (FAL). Inhibition of the Rho pathway by Fasudil completely blocked the beneficial effect of FSS. In a genome-wide gene profiling we identified actin-binding Rho activating protein (Abra), which was highly upregulated in growing collaterals. METHODS AND RESULTS: qRT-PCR and Western blot confirmed highly increased FSS-dependent expression of Abra in growing collaterals. NO blockage by L-NAME abolished FSS-generated Abra expression as well as the whole arteriogenic process. Cell culture studies demonstrated an Abra-triggered proliferation of smooth muscle cells through a mechanism that requires Rho signaling. Local intracollateral adenoviral overexpression of Abra improved collateral conductance by 60% in rabbits compared to the natural response after FAL. In contrast, targeted deletion of Abra in CL57BL/6 mice led to impaired arteriogenesis. CONCLUSIONS: FSS-induced Abra expression during arteriogenesis is triggered by NO and leads to stimulation of collateral growth by smooth muscle cell proliferation.


Asunto(s)
Arterias/crecimiento & desarrollo , Arterias/fisiología , Proteínas de Microfilamentos/metabolismo , Neovascularización Fisiológica , Actinas/metabolismo , Adenoviridae/genética , Animales , Proliferación Celular , Células Cultivadas , Circulación Colateral/fisiología , Técnicas de Transferencia de Gen , Hemorreología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Porcinos , Regulación hacia Arriba , Proteínas de Unión al GTP rho/metabolismo
12.
J Cardiovasc Pharmacol ; 55(2): 153-60, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20173509

RESUMEN

Previous studies showed that targeted endothelial nitric oxide synthase (eNOS) disruption in mice with femoral artery occlusion does not impede and transgenic eNOS overexpression does not stimulate collateral artery growth after femoral artery occlusion, suggesting that nitric oxide from eNOS does not play a role in arteriogenesis. However, pharmacologic nitric oxide synthase inhibition with L-NAME markedly blocks arteriogenesis, suggestive of an important role of nitric oxide. To solve the paradox, we studied targeted deletion of eNOS and of inducible nitric oxide synthase (iNOS) in mice and found that only iNOS knockout could partially inhibit arteriogenesis. However, the combination of eNOS knockout and treatment with the iNOS inhibitor L-NIL completely abolished arteriogenesis. mRNA transcription studies (reverse transcriptase-polymerase chain reaction) performed on collateral arteries of rats showed that eNOS and especially iNOS (but not neural nitric oxide synthase) become upregulated in shear stress-stimulated collateral vessels, which supports the hypothesis that nitric oxide is necessary for arteriogenesis but that iNOS plays an important part. This was strengthened by the observation that the nitric oxide donor DETA NONOate strongly stimulated collateral artery growth, activated perivascular monocytes, and increased proliferation markers. Shear stress-induced nitric oxide may activate the innate immune system and activate iNOS. In conclusion, arteriogenesis is completely dependent on the presence of nitric oxide, a large part of it coming from mononuclear cells.


Asunto(s)
Arterias/efectos de los fármacos , Arterias/crecimiento & desarrollo , Óxido Nítrico/fisiología , Compuestos Nitrosos/farmacología , Animales , Circulación Colateral/efectos de los fármacos , Circulación Colateral/fisiología , Técnicas de Inactivación de Genes , Marcación de Gen , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/fisiología , Conejos , Ratas , Ratas Sprague-Dawley
13.
Cells ; 9(4)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32316253

RESUMEN

Beyond their role in pathogen recognition and the initiation of immune defense, Toll-like receptors (TLRs) are known to be involved in various vascular processes in health and disease. We investigated the potential of the lipopeptide and TLR2/6 ligand macrophage activating protein of 2-kDA (MALP-2) to promote blood flow recovery in mice. Hypercholesterolemic apolipoprotein E (Apoe)-deficient mice were subjected to microsurgical ligation of the femoral artery. MALP-2 significantly improved blood flow recovery at early time points (three and seven days), as assessed by repeated laser speckle imaging, and increased the growth of pre-existing collateral arteries in the upper hind limb, along with intimal endothelial cell proliferation in the collateral wall and pericollateral macrophage accumulation. In addition, MALP-2 increased capillary density in the lower hind limb. MALP-2 enhanced endothelial nitric oxide synthase (eNOS) phosphorylation and nitric oxide (NO) release from endothelial cells and improved the experimental vasorelaxation of mesenteric arteries ex vivo. In vitro, MALP-2 led to the up-regulated expression of major endothelial adhesion molecules as well as their leukocyte integrin receptors and consequently enhanced the endothelial adhesion of leukocytes. Using the experimental approach of femoral artery ligation (FAL), we achieved promising results with MALP-2 to promote peripheral blood flow recovery by collateral artery growth.


Asunto(s)
Circulación Sanguínea/efectos de los fármacos , Arteria Femoral/efectos de los fármacos , Lipopéptidos/farmacología , Macrófagos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 6/metabolismo , Animales , Apolipoproteínas E/deficiencia , Capilares/efectos de los fármacos , Capilares/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Arteria Femoral/diagnóstico por imagen , Arteria Femoral/patología , Arteria Femoral/cirugía , Inmunohistoquímica , Imágenes de Contraste de Punto Láser , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Fosforilación , Vasodilatación/efectos de los fármacos
14.
J Cell Mol Med ; 13(8B): 2613-2621, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19017361

RESUMEN

The development of a collateral circulation (arteriogenesis), bypassing an arterial occlusion, is important for tissue survival, but it remains functionally defective. Micro array data of growing collateral vessels, exposed to chronically elevated fluid shear stress (FSS), showed increased transcription of the transient receptor potential cation channel, subfamily V, member 4 (Trpv4). Thus, the aim of this study was to investigate the role of the shear stress sensitive Trpv4 in transmitting this physical stimulus into an active growth response. qRT-PCR at different time points during the growth of collateral vessels after femoral artery ligature (FAL) in rats showed a strong positive correlation of Trpv4 transcription and the intensity of FSS. An increased protein expression of Trpv4 was localized in the FSS-sensing endothelium by means of confocal immunohistochemistry. Cultured porcine endothelial cells showed a dose-dependent expression of Trpv4 and an increased level of Ki67-positive cells upon treatment with 4alpha-Phorbol 12,13-didecanoate (4alphaPDD), a specific Trpv4 activator. This was also demonstrated by flow culture experiments. These results were confirmed by in vivo application of 4alphaPDD in rabbit hind limb circulation via an osmotic mini-pump after FAL. Trpv4 expression as well as Ki67-positive staining was significantly increased in collateral vessels. Finally, 4alphaPDD treatment after FAL led to a 61% (215.5 ml/min/mmHg versus 350 ml/min/mmHg) recovery of conductance when compared with the non-occluded artery. Cell culture and in vivo studies demonstrate that an FSS- or a 4alphaPDD-induced activation of Trpv4 leads to an active proliferation of vascular cells and finally triggers collateral growth. Trpv4, a well-known FSS-sensitive vasodilator, has hitherto not been implicated in active growth processes of collateral arteries. This new function may lead to new therapeutic strategies for the treatment of arterial occlusive diseases.


Asunto(s)
Circulación Sanguínea/fisiología , Vasos Sanguíneos/crecimiento & desarrollo , Canales Catiónicos TRPV/fisiología , Animales , Vasos Sanguíneos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos
15.
J Clin Invest ; 116(9): 2344-55, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16932810

RESUMEN

Bmx/Etk non-receptor tyrosine protein kinase has been implicated in endothelial cell migration and tube formation in vitro. However, the role of Bmx in vivo is not known. Bmx is highly induced in the vasculature of ischemic hind limbs. We used both mice with a genetic deletion of Bmx (Bmx-KO mice) and transgenic mice expressing a constitutively active form of Bmx under the endothelial Tie-2 enhancer/promoter (Bmx-SK-Tg mice) to study the role of Bmx in ischemia-mediated arteriogenesis/angiogenesis. In response to ischemia, Bmx-KO mice had markedly reduced, whereas Bmx-SK-Tg mice had enhanced, clinical recovery, limb perfusion, and ischemic reserve capacity when compared with nontransgenic control mice. The functional outcomes in these mice were correlated with ischemia-initiated arteriogenesis, capillary formation, and vessel maturation as well as Bmx-dependent expression/activation of TNF receptor 2- and VEGFR2-mediated (TNFR2/VEGFR2-mediated) angiogenic signaling in both hind limb and bone marrow. More importantly, results of bone marrow transplantation studies showed that Bmx in bone marrow-derived cells plays a critical role in the early phase of ischemic tissue remodeling. Our study provides the first demonstration to our knowledge that Bmx in endothelium and bone marrow plays a critical role in arteriogenesis/angiogenesis in vivo and suggests that Bmx may be a novel target for the treatment of vascular diseases such as coronary artery disease and peripheral arterial disease.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Neovascularización Fisiológica , Proteínas Tirosina Quinasas/fisiología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Tirosina Quinasas/genética
16.
Basic Res Cardiol ; 104(1): 5-21, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19101749

RESUMEN

Following an arterial occlusion outward remodeling of pre-existent inter-connecting arterioles occurs by proliferation of vascular smooth muscle and endothelial cells. This is initiated by deformation of the endothelial cells through increased pulsatile fluid shear stress (FSS) caused by the steep pressure gradient between the high pre-occlusive and the very low post-occlusive pressure regions that are interconnected by collateral vessels. Shear stress leads to the activation and expression of all NOS isoforms and NO production, followed by endothelial VEGF secretion, which induces MCP-1 synthesis in endothelium and in the smooth muscle of the media. This leads to attraction and activation of monocytes and T-cells into the adventitial space (peripheral collateral vessels) or attachment of these cells to the endothelium (coronary collaterals). Mononuclear cells produce proteases and growth factors to digest the extra-cellular scaffold and allow motility and provide space for the new cells. They also produce NO from iNOS, which is essential for arteriogenesis. The bulk of new tissue production is carried by the smooth muscles of the media, which transform their phenotype from a contractile into a synthetic and proliferative one. Important roles are played by actin binding proteins like ABRA, cofilin, and thymosin beta 4 which determine actin polymerization and maturation. Integrins and connexins are markedly up-regulated. A key role in this concerted action which leads to a 2-to-20 fold increase in vascular diameter, depending on species size (mouse versus human) are the transcription factors AP-1, egr-1, carp, ets, by the Rho pathway and by the Mitogen Activated Kinases ERK-1 and -2. In spite of the enormous increase in tissue mass (up to 50-fold) the degree of functional restoration of blood flow capacity is incomplete and ends at 30% of maximal conductance (coronary) and 40% in the vascular periphery. The process of arteriogenesis can be drastically stimulated by increases in FSS (arterio-venous fistulas) and can be completely blocked by inhibition of NO production, by pharmacological blockade of VEGF-A and by the inhibition of the Rho-pathway. Pharmacological stimulation of arteriogenesis, important for the treatment of arterial occlusive diseases, seems feasible with NO donors.


Asunto(s)
Arteriolas/citología , Arteriolas/fisiología , Circulación Colateral/fisiología , Animales , División Celular , Endotelio Vascular/citología , Humanos , Isquemia/fisiopatología , Músculo Liso Vascular/citología
17.
J Vasc Res ; 46(4): 365-74, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19142016

RESUMEN

BACKGROUND/AIMS: Collateral vessels restore only about 40% of the maximum dilatory reserve after femoral artery occlusion, whereas complete normalization is reached by increased fluid shear stress (FSS). We studied the role of known potent angiogenic growth factors (separately or in combination) in arteriogenesis by determining their expression in FSS-stimulated collaterals and close-to-collateral infusion of growth factor peptides in a rabbit model of femoral artery occlusion. METHODS: Values of maximum collateral conductance (C(max)) and post mortem angiograms were compared to those achievable by high FSS. mRNA levels of growth factor ligands and receptors were determined in FSS-stimulated collaterals. RESULTS: Seven days after vessel occlusion, FSS-stimulated legs showed a C(max) not significantly different from that of not occluded femoral arteries. Arteriogenesis was significantly less enhanced after growth factor treatment (MCP-1 86%, Ad5.1-FGF-4 75%, bFGF 72%, PDGF 64%, VEGF 50% of C(max) after FSS stimulation). RT-PCR showed no differential expression of FGF receptors, but an up-regulation of VEGF-receptor-2. CONCLUSION: The most potent known angiogenic growth factors at high pharmacological doses reach only a fraction of the maximum conductance obtained by high FSS. Arteriogenesis differs from angiogenesis, so the main focus to markedly improve arteriogenesis should be put on the underlying mechanisms of shear stress.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Arteriopatías Oclusivas/metabolismo , Circulación Colateral , Arteria Femoral/metabolismo , Neovascularización Fisiológica , Proteínas Angiogénicas/administración & dosificación , Proteínas Angiogénicas/genética , Animales , Arteriopatías Oclusivas/genética , Arteriopatías Oclusivas/fisiopatología , Derivación Arteriovenosa Quirúrgica , Quimiocina CCL2/metabolismo , Circulación Colateral/genética , Modelos Animales de Enfermedad , Arteria Femoral/diagnóstico por imagen , Arteria Femoral/fisiopatología , Arteria Femoral/cirugía , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Hemorreología , Bombas de Infusión , Ligadura , Masculino , Neovascularización Fisiológica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , ARN Mensajero/metabolismo , Conejos , Radiografía , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Estrés Mecánico , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Mol Cell Biochem ; 322(1-2): 15-23, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18979232

RESUMEN

Arteriogenesis or collateral growth is able to compensate for the stenosis of major arteries. Using differential display RT-PCR on growing and quiescent collateral arteries in a rabbit femoral artery ligation model, we cloned the rabbit full-length cDNA of osteoglycin/mimecan. Osteoglycin was present in the adventitia of collateral arteries as a glycosylated protein without keratan sulfate side chains, mainly produced by smooth muscle cells (SMCs) and perivascular fibroblasts. Northern blot, Western blot, and immunohistochemistry confirmed a collateral artery-specific downregulation of osteoglycin from 6 h to 3 weeks after the onset of arteriogenesis. Treatment of primary SMCs with the arteriogenic protein fibroblast growth factor-2 (FGF-2) resulted in a similar reduction of osteoglycin expression as observed in vivo. Application of the FGF-2 inhibitor polyanethole sulfonic acid (PAS) blocked the downregulation of osteoglycin and interfered with arteriogenesis. From our study we conclude that downregulation of osteoglycin is a fundamental requirement for proper arteriogenesis.


Asunto(s)
Arteria Femoral/crecimiento & desarrollo , Proteoglicanos/metabolismo , Secuencia de Aminoácidos , Animales , Arterias/crecimiento & desarrollo , Secuencia de Bases , ADN Complementario/química , Regulación hacia Abajo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Modelos Animales , Datos de Secuencia Molecular , Músculo Liso Vascular/metabolismo , Proteoglicanos/química , Proteoglicanos/genética , ARN Mensajero/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Mol Cell Biochem ; 322(1-2): 161-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18998200

RESUMEN

Migration and proliferation of smooth muscle cells (SMC) are important events during arteriogenesis, but the underlying mechanism is still only partially understood. The present study investigates the expression of integrins alpha 5 beta 1 and v beta 3 as well as focal adhesion kinase (FAK) and phosphorylated FAK (pY397), key mediators for cell migration and proliferation, in collateral vessels (CV) in rabbit hind limbs induced by femoral ligation or an arteriovenous (AV) shunt created between the distal femoral artery stump and the accompanying femoral vein by confocal immunofluorescence. In addition, the effect of the extracellular matrix components fibronectin (FN), laminin (LN), and Matrigel on expression of these focal adhesion molecules proliferation was studied in cultured SMCs. We found that: (1) in normal vessels (NV), both integrins alpha 5 beta 1 and alpha v beta 3 were mainly expressed in endothelial cells, very weak in smooth muscle cells (SMC); (2) in CVs, both alpha 5 beta 1 and alpha v beta 3 were significantly upregulated (P < 0.05); this was more evident in the shunt-side CVs, 1.5 and 1.3 times higher than that in the ligation side, respectively; (3) FAK and FAK(py397) were expressed in NVs and CVs in a similar profile as was alpha 5 beta 1 and alpha v beta 3; (4) in vitro SMCs cultured on fibronectin (overexpressed in collaterals) expressed higher levels of FAK, FAK (pY397), alpha 5 beta 1, and alpha v beta 3 than on laminin, whereas SMCs growing inside Matrigel expressed little of these proteins and showed no proliferation. In conclusion, our data demonstrate for the first time that the integrin-FAK signaling axis is activated in collateral vessels and that altered expression of FN and LN may play a crucial role in mediating the integrin-FAK signaling pathway activation. These findings explain a large part of the positive remodeling that collateral vessels undergo under the influence of high fluid shear stress.


Asunto(s)
Arterias/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Animales , Arterias/crecimiento & desarrollo , Proliferación Celular , Células Cultivadas , Arteria Femoral/citología , Arteria Femoral/metabolismo , Inmunohistoquímica , Microscopía Electrónica , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Conejos , Remodelación Ventricular
20.
Circ Res ; 101(1): 88-96, 2007 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-17540977

RESUMEN

Angiopoietins play important roles in the formation of neovessels and complex vascular networks. Angiopoietin (Ang)-1 and Ang-2 belong to a family of growth factors that display opposing effects on the activation of Tie2 (tyrosine kinase with immunoglobulin and epidermal growth factor homology domain 2). Endothelial Ang-2 expression is associated with vessel destabilization and regulates a balance between vascular regression and growth. To elucidate, in particular, the role of Ang-2 after arterial artery occlusion in the mouse limb, we applied a transgenic animal model with targeted Ang-2 expression in endothelial cells. We show here that restoration of blood flow in Ang-2:Tie1 transgenic mice is dramatically impaired when Ang-2 expression is induced in the vasculature. The defective restoration of perfusion in Ang-2 transgenic mice is evidenced by reduced collateral artery growth, which typically occurs to compensate for flow deficits after occlusion of the large conductance artery. Furthermore, reduced movement capacities and higher incidents of necrosis are consequently observed in the transgenic limbs as compared with controls. Mechanistically, the observed effects are attributed to defective smooth muscle cell recruitment in Ang-2 transgenic mice. Moreover, distinct Ang-2 levels in the genetically modified animals clearly correlated with the magnitude of reduced perfusion. In conclusion, our studies define Ang-2 as an important molecule for the progression of collateral artery growth and angiogenesis during ischemia and suggest precise Ang-2 dosage activities to accomplish blood vessel growth.


Asunto(s)
Angiopoyetina 2/fisiología , Miembro Posterior/irrigación sanguínea , Miembro Posterior/metabolismo , Isquemia/metabolismo , Isquemia/fisiopatología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Angiopoyetina 2/genética , Animales , Arteriopatías Oclusivas/genética , Arteriopatías Oclusivas/metabolismo , Arteriopatías Oclusivas/fisiopatología , Velocidad del Flujo Sanguíneo/genética , Isquemia/genética , Ratones , Ratones Transgénicos , Neovascularización Patológica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA