Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(4): 104594, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36898577

RESUMEN

Cluster of differentiation 47 (CD47) plays an important role in the pathophysiology of various diseases including atherosclerosis but its role in neointimal hyperplasia which contributes to restenosis has not been studied. Using molecular approaches in combination with a mouse vascular endothelial denudation model, we studied the role of CD47 in injury-induced neointimal hyperplasia. We determined that thrombin-induced CD47 expression both in human aortic smooth muscle cells (HASMCs) and mouse aortic smooth muscle cells. In exploring the mechanisms, we found that the protease-activated receptor 1-Gα protein q/11 (Gαq/11)-phospholipase Cß3-nuclear factor of activated T cells c1 signaling axis regulates thrombin-induced CD47 expression in HASMCs. Depletion of CD47 levels using its siRNA or interference of its function by its blocking antibody (bAb) blunted thrombin-induced migration and proliferation of HASMCs and mouse aortic smooth muscle cells. In addition, we found that thrombin-induced HASMC migration requires CD47 interaction with integrin ß3. On the other hand, thrombin-induced HASMC proliferation was dependent on CD47's role in nuclear export and degradation of cyclin-dependent kinase-interacting protein 1. In addition, suppression of CD47 function by its bAb rescued HASMC efferocytosis from inhibition by thrombin. We also found that vascular injury induces CD47 expression in intimal SMCs and that inhibition of CD47 function by its bAb, while alleviating injury-induced inhibition of SMC efferocytosis, attenuated SMC migration, and proliferation resulting in reduced neointima formation. Thus, these findings reveal a pathological role for CD47 in neointimal hyperplasia.


Asunto(s)
Antígeno CD47 , Reestenosis Coronaria , Miocitos del Músculo Liso , Animales , Humanos , Ratones , Antígeno CD47/antagonistas & inhibidores , Antígeno CD47/genética , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Hiperplasia/metabolismo , Hiperplasia/fisiopatología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Neointima/fisiopatología , Trombina/metabolismo , Lesiones del Sistema Vascular/fisiopatología , Regulación de la Expresión Génica/genética , Reestenosis Coronaria/fisiopatología
2.
Biochem Soc Trans ; 49(1): 353-363, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33616623

RESUMEN

MicroRNAs are short, non-coding RNAs that target messenger RNAs for degradation. miR-145 is a vascular-enriched microRNA that is important for smooth muscle cell (SMC) differentiation. Under healthy circumstances, SMC exist in a contractile, differentiated phenotype promoted by miR-145. In cases of disease or injury, SMC can undergo reversible dedifferentiation into a synthetic phenotype, accompanied by inhibition of miR-145 expression. Vascular disorders such as atherosclerosis and neointimal hyperplasia are characterised by aberrant phenotypic switching in SMC. This review will summarise the physiological roles of miR-145 in vascular SMC, including the molecular regulation of differentiation, proliferation and migration. Furthermore, it will discuss the different ways in which miR-145 can be dysregulated and the downstream impact this has on the progression of vascular pathologies. Finally, it will discuss whether miR-145 may be suitable for use as a biomarker of vascular disease.


Asunto(s)
Enfermedades Cardiovasculares/genética , MicroARNs/fisiología , Miocitos del Músculo Liso/fisiología , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Enfermedades Cardiovasculares/fisiopatología , Salud , Humanos , MicroARNs/genética , Contracción Muscular/genética , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/fisiopatología , Neointima/metabolismo , Neointima/fisiopatología
3.
Cardiovasc Drugs Ther ; 35(5): 915-925, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33394362

RESUMEN

PURPOSE: Anti-proliferative drugs released from drug-eluting stents delay cell coverage and vascular healing, which increases the risk of late stent thrombosis. We assessed the potential effects of systemic methotrexate (MTX) on cell coverage, vascular healing and inflammation activation in vivo and in vitro. METHODS: We applied MTX in the right common carotid artery in a rabbit stenting model to determine the impact on cell coverage and inflammation activation using a serial optical coherence tomography (OCT) analysis and elucidated the molecular mechanism of MTX in human umbilical vein endothelial cells (HUVECs). RESULTS: Low-dose MTX promoted the development of cell coverage and vascular healing, which was associated with fewer uncovered struts (%) and cross-sections with any uncovered struts (%) at 4 weeks of stenting. The MTX group also exhibited lower rates of heterogeneity, microvessels and per-strut low-signal-intensity layers, indicating neointimal instability at 12 weeks of stenting. In vitro, low-dose MTX strongly inhibited HUVEC apoptosis, promoted proliferation and inhibited inflammatory activation by targeting the phosphoinositide 3-kinase (PI3K)/AKT signalling pathway. CONCLUSION: Low-dose MTX may be a key means of promoting early cell coverage via the inhibition of the inflammatory response and stability of neointima by targeting inflammatory pathways after stent implantation.


Asunto(s)
Arteria Carótida Común/efectos de los fármacos , Stents Liberadores de Fármacos/efectos adversos , Mediadores de Inflamación/metabolismo , Metotrexato/farmacología , Neointima/fisiopatología , Quinasa de Linfoma Anaplásico/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Conejos , Distribución Aleatoria
4.
Arterioscler Thromb Vasc Biol ; 39(3): e91-e105, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30651001

RESUMEN

Objective- Vascular smooth muscle cells (VSMCs) phenotype modulation is critical for the resolution of vascular injury. Genetic and pharmacological inhibition of PI3Kγ (phosphoinositide 3-kinase γ) exerts anti-inflammatory and protective effects in multiple cardiovascular diseases. This study investigated the role of PI3Kγ and its downstream effector molecules in the regulation of VSMC phenotypic modulation and neointimal formation in response to vascular injury. Approach and Results- Increased expression of PI3Kγ was found in injured vessel wall as well in cultured, serum-activated wild-type VSMCs, accompanied by a reduction in the expression of calponin and SM22α, 2 differentiation markers of VSMCs. However, the injury-induced downregulation of calponin and SM22α was profoundly attenuated in PI3Kγ-/- mice. Pharmacological inhibition and short hairpin RNA knockdown of PI3Kγ (PI3Kγ-KD) markedly attenuated YAP (Yes-associated protein) expression and CREB (cyclic AMP-response element binding protein) activation but improved the downregulation of differentiation genes in cultured VSMCs accompanied by reduced cell proliferation and migration. Mechanistically, activated CREB upregulated YAP transcriptional expression through binding to its promoter. Ectopic expression of YAP strikingly repressed the expression of differentiation genes even in PI3Kγ-KD VSMCs. Moreover, established carotid artery ligation and chimeric mice models demonstrate that deletion of PI3Kγ in naïve PI3Kγ-/- mice as well as in chimeric mice lacking PI3Kγ either in bone marrow or vascular wall significantly reduced neointimal formation after injury. Conclusions- PI3Kγ controls phenotypic modulation of VSMCs by regulating transcription factor CREB activation and YAP expression. Modulating PI3Kγ signaling on local vascular wall may represent a new therapeutic approach to treat proliferative vascular disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Ciclo Celular/fisiología , Fosfatidilinositol 3-Quinasa Clase Ib/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Neointima/fisiopatología , Transducción de Señal/fisiología , Animales , Arteria Carótida Común , Movimiento Celular , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/patología , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/genética , Quimera por Radiación , Remodelación Vascular , Proteínas Señalizadoras YAP
5.
Circ J ; 83(7): 1472-1479, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31061352

RESUMEN

BACKGROUND: Carotid angioplasty and stenting (CAS) is emerging as an alternative treatment for carotid stenosis, but neointimal hyperplasia (NIH) remains a drawback of this treatment strategy. This study aimed to evaluate the effect of variations of carotid bifurcation geometry on local hemodynamics and NIH.Methods and Results:Hemodynamic and geometric effects on NIH were compared between 2 groups, by performing computational fluid dynamics (CFD) simulations both on synthetic models and patient-specific models. In the idealized models, multiple regression analysis revealed a significant negative relationship between internal carotid artery (ICA) angle and the local hemodynamics. In the patient-derived models, which were reconstructed from digital subtraction angiography (DSA) of 25 patients with bilateral CAS, a low time-average wall shear stress (TAWSS) and a high oscillatory shear index (OSI) were often found at the location of NIH. Larger difference values of the OSI percentage area (10.56±20.798% vs. -5.87±18.259%, P=0.048) and ECA/CCA diameter ratio (5.64±12.751% vs. -3.59±8.697%, P=0.047) were detected in the NIH-asymmetric group than in the NIH-symmetric group. CONCLUSIONS: Changes in carotid bifurcation geometry can make apparent differences in hemodynamic distribution and lead to bilateral NIH asymmetry. It may therefore be reasonable to consider certain geometric variations as potential local risk factors for NIH.


Asunto(s)
Arterias Carótidas , Estenosis Carotídea , Hidrodinámica , Modelos Cardiovasculares , Neointima , Stents , Anciano , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Arterias Carótidas/cirugía , Estenosis Carotídea/patología , Estenosis Carotídea/fisiopatología , Estenosis Carotídea/cirugía , Femenino , Estudios de Seguimiento , Humanos , Hiperplasia/patología , Hiperplasia/fisiopatología , Masculino , Persona de Mediana Edad , Neointima/patología , Neointima/fisiopatología
6.
Int J Med Sci ; 16(1): 106-114, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30662334

RESUMEN

This study shows that microRNA-320 (miR-320) is associated with many important cell functions, including cell differentiation, proliferation, migration, and apoptosis. However, the role of miR-320 in vascular smooth muscle cells (VSMCs) and proliferative vascular diseases is still completely unclear. In our study, we found that the expression of miR-320 in human VSMCs after PDGF stimulation was significantly down-regulated in time- and dose-dependent manner. Function analyses identified that miR-320 could inhibit the proliferation and migration of VSMCs in both basal and PDGF-stimulated conditions. Furthermore, Neuropilin 1 (NRP1) was demonstrated as a direct target of miR-320 in Luciferase reporter assays and miR-320 overexpression inhibited the expression of NRP1 with or without PDGF treatment. Finally, miR-320 was markedly decreased in mice carotid arteries after ligated injury, while the restoration of miR-320 via Ad-miR-320 attenuated neointimal hyperplasia by declining the NRP1 expression. The results confirmed that miR-320 regulated proliferation and migration of VSMCs and neointimal formation by targeting NRP1. These novel findings implied that the regulation of NRP1 expression by miR-320 has important significance in the early diagnosis and treatment of proliferation vascular diseases.


Asunto(s)
MicroARNs/metabolismo , Músculo Liso Vascular/fisiología , Neointima/fisiopatología , Neuropilina-1/metabolismo , Animales , Aorta/fisiología , Arterias Carótidas/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Regulación hacia Abajo , Humanos , Masculino , Ratones , Factor de Crecimiento Derivado de Plaquetas/metabolismo
7.
J Sci Food Agric ; 99(9): 4397-4406, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30861122

RESUMEN

BACKGROUND: Compounds of the inner shell of chestnut (Castanea crenata) have diverse biological activities, including anti-cancer and anti-oxidant activities. Here we explored the effects of an extract of chestnut inner shells and of its bioactive component scoparone on vascular smooth muscle cell migration and vessel damage. RESULTS: The ethanol extract of chestnut inner shells, containing 11 major compounds, inhibited platelet-derived growth factor (PDGF)-BB-induced migration of rat aortic smooth muscle cells (RASMCs). Among these compounds, scoparone (6,7-dimethoxycoumarin) suppressed RASMC migration and wound healing in response to PDGF-BB but did not affect RASMC proliferation. In RASMCs, scoparone inhibited the PDGF-BB-induced rat aortic sprout outgrowth and attenuated the PDGF-BB-mediated increase in phosphorylation of mitogen-activated protein kinases (MAPKs), p38 MAPK and extracellular signal-regulated kinase 1/2. The in vivo administration of scoparone resulted in the attenuation of neointima formation in balloon-injured carotid arteries of rats. CONCLUSION: These findings demonstrate that scoparone, found in chestnut inner shells, may inhibit cell migration through suppression of the phosphorylation of MAPKs in PDGF-BB-treated RASMCs, probably contributing to the reduction of neointimal hyperplasia induced after vascular injury. Therefore, scoparone and chestnut inner shell may be a potential agent or functional food, respectively, for the prevention of vascular disorders such as vascular restenosis or atherosclerosis. © 2019 Society of Chemical Industry.


Asunto(s)
Becaplermina/metabolismo , Cumarinas/administración & dosificación , Fagaceae/química , Hiperplasia/tratamiento farmacológico , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Neointima/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cumarinas/química , Humanos , Hiperplasia/fisiopatología , Masculino , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Neointima/fisiopatología , Nueces/química , Extractos Vegetales/química , Ratas , Ratas Sprague-Dawley
8.
Rev Port Cir Cardiotorac Vasc ; 26(3): 213-217, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31734974

RESUMEN

Neointimal hyperplasia is a physiologic healing response to injury to the blood vessel wall, involving all the three arterial layers and it occurs in the presence of internal (endovascular) or external (surgical) injury. It is a highly complex process involving several tissues (perivascular, vessel wall, and blood) and numerous cell lineages with multiple molecular signaling networks. So, there is a number of possible targets for inhibition of this process. There are known risk factors for Intimal Hyperplasia, such as diabetes, female gender, presence of systemic inflammation, type of arteries treated, types of surgical and endovascular materials, presence of turbulent flow and genetic status. The present paper discusses the pathophysiology of neointimal hyperplasia and the strategies to prevention and treatment of it.


Asunto(s)
Arterias/fisiopatología , Neointima/fisiopatología , Neointima/terapia , Arterias/patología , Femenino , Humanos , Hiperplasia , Neointima/etiología , Neointima/patología
9.
J Mol Cell Cardiol ; 121: 145-154, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30003882

RESUMEN

BACKGROUND: Antiproliferative drugs in drug eluting stents (DES) are associated with complications due to impaired re-endothelialization. Additionally, adventitial neovascularization has been suggested to contribute to in-stent restenosis (ISR). Since Vascular Endothelial Growth Factors (VEGFs) are the key mediators of angiogenesis, we investigated feasibility and efficacy of local gene therapy for ISR utilizing soluble decoy VEGF receptors to reduce biological activity of adventitial VEGFs. METHOD: Sixty-nine adult WHHL rabbit aortas were subjected to endothelial denudation. Six weeks later catheter-mediated local intramural infusion of 1.5x10e10 pfu adenoviruses encoding soluble VEGF Receptor-1 (sVEGFR1), sVEGFR2, sVEGFR3 or control LacZ and bare metal stent implantation were performed in the same aortic segment. Marker protein expression was assessed at 6d in LacZ cohort. Immunohistochemistry, morphometrical analyses and angiography were performed at d14, d42 and d90. RESULTS: Transgene expression was localized to adventitia. All decoy receptors reduced the size of vasa-vasorum at 14d, AdsVEGFR2 animals also had reduced density of adventitial vasa-vasorum, whereas AdsVEGFR3 increased the density of vasa-vasorum. At d42, AdsVEGFR1 and AdsVEGFR2 reduced ISR (15.7 ±â€¯6.9% stenosis, P < 0.01 and 16.5 ±â€¯2.7%, P < 0.05, respectively) vs. controls (28.3 ±â€¯7.6%). Moreover, AdsVEGFR-3 treatment led to a non-significant trend in the reduction of adventitial lymphatics at all time points and these animals had significantly more advanced neointimal atherosclerosis at 14d and 42d vs. control animals. CONCLUSIONS: Targeting adventitial neovascularization using sVEGFR1 and sVEGFR2 is a novel strategy to reduce ISR. The therapeutic effects dissipate at late follow up following short expression profile of adenoviral vectors. However, inhibition of VEGFR3 signaling accelerates neoatherosclerosis.


Asunto(s)
Constricción Patológica/terapia , Reestenosis Coronaria/terapia , Terapia Genética , Neointima/terapia , Neovascularización Patológica/tratamiento farmacológico , Adventicia/fisiopatología , Animales , Aorta/fisiopatología , Constricción Patológica/genética , Constricción Patológica/fisiopatología , Reestenosis Coronaria/genética , Reestenosis Coronaria/fisiopatología , Stents Liberadores de Fármacos , Endotelio/citología , Endotelio/efectos de los fármacos , Endotelio/crecimiento & desarrollo , Endotelio Vascular/fisiopatología , Humanos , Neointima/genética , Neointima/fisiopatología , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Conejos , Vasa Vasorum/fisiopatología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Factores de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular/uso terapéutico
10.
Circ Res ; 119(12): 1313-1323, 2016 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-27682618

RESUMEN

RATIONALE: Vascular smooth muscle cell (VSMC) accumulation is a hallmark of atherosclerosis and vascular injury. However, fundamental aspects of proliferation and the phenotypic changes within individual VSMCs, which underlie vascular disease, remain unresolved. In particular, it is not known whether all VSMCs proliferate and display plasticity or whether individual cells can switch to multiple phenotypes. OBJECTIVE: To assess whether proliferation and plasticity in disease is a general characteristic of VSMCs or a feature of a subset of cells. METHODS AND RESULTS: Using multicolor lineage labeling, we demonstrate that VSMCs in injury-induced neointimal lesions and in atherosclerotic plaques are oligoclonal, derived from few expanding cells. Lineage tracing also revealed that the progeny of individual VSMCs contributes to both alpha smooth muscle actin (aSma)-positive fibrous cap and Mac3-expressing macrophage-like plaque core cells. Costaining for phenotypic markers further identified a double-positive aSma+ Mac3+ cell population, which is specific to VSMC-derived plaque cells. In contrast, VSMC-derived cells generating the neointima after vascular injury generally retained the expression of VSMC markers and the upregulation of Mac3 was less pronounced. Monochromatic regions in atherosclerotic plaques and injury-induced neointima did not contain VSMC-derived cells expressing a different fluorescent reporter protein, suggesting that proliferation-independent VSMC migration does not make a major contribution to VSMC accumulation in vascular disease. CONCLUSIONS: We demonstrate that extensive proliferation of a low proportion of highly plastic VSMCs results in the observed VSMC accumulation after injury and in atherosclerotic plaques. Therapeutic targeting of these hyperproliferating VSMCs might effectively reduce vascular disease without affecting vascular integrity.


Asunto(s)
Aterosclerosis/fisiopatología , Proliferación Celular/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Neointima/fisiopatología , Lesiones del Sistema Vascular/fisiopatología , Animales , Aterosclerosis/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/citología , Neointima/patología , Lesiones del Sistema Vascular/patología
11.
Arterioscler Thromb Vasc Biol ; 37(10): 1944-1955, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28798140

RESUMEN

OBJECTIVE: The role of hemoglobin and myoglobin in the cardiovascular system is well established, yet other globins in this context are poorly characterized. Here, we examined the expression and function of cytoglobin (CYGB) during vascular injury. APPROACH AND RESULTS: We characterized CYGB content in intact vessels and primary vascular smooth muscle (VSM) cells and used 2 different vascular injury models to examine the functional significance of CYGB in vivo. We found that CYGB was strongly expressed in medial arterial VSM and human veins. In vitro and in vivo studies indicated that CYGB was lost after VSM cell dedifferentiation. In the rat balloon angioplasty model, site-targeted delivery of adenovirus encoding shRNA specific for CYGB prevented its reexpression and decreased neointima formation. Similarly, 4 weeks after complete ligation of the left common carotid, Cygb knockout mice displayed little to no evidence of neointimal hyperplasia in contrast to their wild-type littermates. Mechanistic studies in the rat indicated that this was primarily associated with increased medial cell loss, terminal uridine nick-end labeling staining, and caspase-3 activation, all indicative of prolonged apoptosis. In vitro, CYGB could be reexpressed after VSM stimulation with cytokines and hypoxia and loss of CYGB sensitized human and rat aortic VSM cells to apoptosis. This was reversed after antioxidant treatment or NOS2 (nitric oxide synthase 2) inhibition. CONCLUSIONS: These results indicate that CYGB is expressed in vessels primarily in differentiated medial VSM cells where it regulates neointima formation and inhibits apoptosis after injury.


Asunto(s)
Apoptosis , Globinas/fisiología , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiopatología , Remodelación Vascular/fisiología , Animales , Caspasa 3/metabolismo , Diferenciación Celular , Citoglobina , Regulación hacia Abajo , Activación Enzimática , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Neointima/fisiopatología , Óxido Nítrico Sintasa de Tipo II/toxicidad , Oxidación-Reducción , Ratas
12.
Circ Res ; 116(7): 1120-32, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25608528

RESUMEN

RATIONALE: Neointimal hyperplasia characterized by abnormal accumulation of vascular smooth muscle cells (SMCs) is a hallmark of occlusive disorders such as atherosclerosis, postangioplasty restenosis, vein graft stenosis, and allograft vasculopathy. Cyclic nucleotides are vital in SMC proliferation and migration, which are regulated by cyclic nucleotide phosphodiesterases (PDEs). OBJECTIVE: Our goal is to understand the regulation and function of PDEs in SMC pathogenesis of vascular diseases. METHODS AND RESULTS: We performed screening for genes differentially expressed in normal contractile versus proliferating synthetic SMCs. We observed that PDE1C expression was low in contractile SMCs but drastically elevated in synthetic SMCs in vitro and in various mouse vascular injury models in vivo. In addition, PDE1C was highly induced in neointimal SMCs of human coronary arteries. More importantly, injury-induced neointimal formation was significantly attenuated by PDE1C deficiency or PDE1 inhibition in vivo. PDE1 inhibition suppressed vascular remodeling of human saphenous vein explants ex vivo. In cultured SMCs, PDE1C deficiency or PDE1 inhibition attenuated SMC proliferation and migration. Mechanistic studies revealed that PDE1C plays a critical role in regulating the stability of growth factor receptors, such as PDGF receptor ß (PDGFRß) known to be important in pathological vascular remodeling. PDE1C interacts with low-density lipoprotein receptor-related protein-1 and PDGFRß, thus regulating PDGFRß endocytosis and lysosome-dependent degradation in an low-density lipoprotein receptor-related protein-1-dependent manner. A transmembrane adenylyl cyclase cAMP-dependent protein kinase cascade modulated by PDE1C is critical in regulating PDGFRß degradation. CONCLUSIONS: These findings demonstrated that PDE1C is an important regulator of SMC proliferation, migration, and neointimal hyperplasia, in part through modulating endosome/lysosome-dependent PDGFRß protein degradation via low-density lipoprotein receptor-related protein-1.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/enzimología , Neointima/enzimología , Animales , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , División Celular , Movimiento Celular , Células Cultivadas , AMP Cíclico/fisiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/deficiencia , Endocitosis/fisiología , Inducción Enzimática , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Lisosomas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Miocitos del Músculo Liso/citología , Neointima/fisiopatología , Mapeo de Interacción de Proteínas , Estabilidad Proteica , Proteolisis , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología
13.
Cell Biol Int ; 41(6): 586-598, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28328100

RESUMEN

At the early stage of atherosclerosis, neointima is formed due to the migration of vascular smooth muscle cells (VSMCs) from the media to the intima. VSMCs are surrounded by highly adhesive 3D matrices. They take specific strategies to cross various 3D matrices in the media, including heterogeneous collagen and mechanically strong basement membrane. Migration of VSMCs is potentially caused by biomechanical mechanism. Most in vitro studies focus on cell migration on 2D substrates in response to biochemical factors. How the cells move through 3D matrices under the action of mechanosensing machineries remains unexplored. In this review, we propose that several interesting tension-dependent machineries act as "tractor"-posterior myosin II accumulation, and "wrecker"-anterior podosome maintaining, to power VSMCs ahead. VSMCs embedded in 3D matrices may accumulate a minor myosin II isoform, myosin IIB, at the cell rear. Anisotropic myosin IIB distribution creates cell rear, polarizes cell body, pushes the nucleus and reshapes the cell body, and cooperates with a uniformly distributed myosin IIA to propel the cell forward. On the other hand, matrix digestion by podosome further promote the migration when the matrix becomes denser. Actomyosin tension activates Src to induce podosome in soft 3D matrices and retain the podosome integrity to steadily digest the matrix.


Asunto(s)
Polaridad Celular/fisiología , Mecanotransducción Celular/fisiología , Músculo Liso Vascular/fisiología , Animales , Aterosclerosis/fisiopatología , Línea Celular , Movimiento Celular/fisiología , Células Cultivadas , Colágeno , Humanos , Miocitos del Músculo Liso , Neointima/fisiopatología , Miosina Tipo IIB no Muscular/metabolismo , Miosina Tipo IIB no Muscular/fisiología
14.
Biochem Biophys Res Commun ; 477(4): 774-780, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27387235

RESUMEN

Phenotype switching of vascular smooth muscle cells (VSMC) from the contractile type to the synthetic type is a hallmark of vascular disorders such as atherosclerosis and restenosis after angioplasty. Inward rectifier K(+) channel 2.1 (Kir2.1) has been identified in VSMC. However, whether it plays a functional role in regulating cellular transformation remains obscure. In this study, we evaluated the role of Kir2.1 on VSMC proliferation, migration, phenotype switching, and post-injury carotid neointimal formation. Kir2.1 knockdown significantly suppressed platelet-derived growth factor BB-stimulated rat vascular smooth muscle cells (rat-VSMC) proliferation and migration. Deficiency in Kir2.1 contributed to the restoration of smooth muscle α-actin, smooth muscle 22α, and calponin and to a reduction in osteopontin expression in rat-VSMC. Moreover, the in vivo study showed that rat-VSMC switched to proliferative phenotypes and that knockdown of Kir2.1 significantly inhibited neointimal formation after rat carotid injury. Kir2.1 may be a potential therapeutic target in the treatment of cardiovascular diseases, such as atherosclerosis and restenosis following percutaneous coronary intervention.


Asunto(s)
Traumatismos de las Arterias Carótidas/patología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Miocitos del Músculo Liso/fisiología , Neointima/fisiopatología , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Traumatismos de las Arterias Carótidas/metabolismo , Células Cultivadas , Masculino , Miocitos del Músculo Liso/patología , Neointima/patología , Ratas , Ratas Sprague-Dawley , Regeneración/fisiología
15.
Biochem Biophys Res Commun ; 479(2): 125-131, 2016 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-27404127

RESUMEN

Growth hormone secretagogue receptor (GHSR) is involved in appetite regulation and energy homeostasis. In the present study, we examined the role of GHSR in neointimal formation following vascular injury. In the mouse model of femoral artery wire injury, we found that vessel intima-to-media ratio was significantly reduced in GHSR deficiency (GHSR-/-) mice compared with that in wild-type mice. Immunohistochemical staining showed that the smooth muscle cell (SMCs) in the neointima were significantly decreased in the injured arteries of GHSR-/- mice which was associated with decreased SMC proliferation and migration. Furthermore, immunoblotting demonstrated that, in cultured rat aortic SMCs, small interfering RNA-mediated GHSR knockdown suppressed the activation of Akt and ERK1/2 signaling pathway. These findings suggested a novel role of GHSR in neointimal formation likely via promoting the proliferation and migration of SMCs involving Akt and ERK1/2 signaling. Therefore, GHSR may be a potential therapeutic target in restenosis and vascular remodeling.


Asunto(s)
Arteria Femoral/lesiones , Arteria Femoral/fisiopatología , Neointima/fisiopatología , Receptores de Ghrelina/deficiencia , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Arteria Femoral/metabolismo , Immunoblotting , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Ratas Sprague-Dawley , Receptores de Ghrelina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
FASEB J ; 29(1): 141-51, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25300621

RESUMEN

Obesity is highly linked with the development of vascular diseases. Omentin is a circulating adipokine that is downregulated in patients with cardiovascular diseases. In this study, we investigated the role of omentin in regulation of vascular remodeling in response to injury. Wild-type (WT) mice were treated intravenously with adenoviral vectors encoding human omentin (Ad-OMT) or control ß-gal and subjected to arterial wire injury. Ad-OMT treatment reduced the neointimal thickening and the frequencies of bromodeoxyuridine-positive proliferating cells in injured arteries. Treatment of vascular smooth muscle cells (VSMCs) with human omentin protein at a physiologic concentration led to suppression of growth and ERK phosphorylation after stimulation with various growth factors. Omentin stimulated AMPK signaling in VSMCs, and blockade of AMPK reversed omentin-mediated inhibition of VSMC growth and ERK phosphorylation. Furthermore, fat-specific human omentin transgenic (OMT-TG) mice exhibited reduced neointimal thickening and vascular cell growth following vascular injury. AMPK activation was enhanced in injured arteries in OMT-TG mice, and administration of AMPK inhibitor reversed the reduction of neointimal hyperplasia in OMT-TG mice. These data indicate that omentin attenuates neointimal formation after arterial injury and suppresses VSMC growth through AMPK-dependent mechanisms. Thus, omentin can represent a novel target molecule for the prevention of vascular disorders.


Asunto(s)
Citocinas/fisiología , Arteria Femoral/lesiones , Lectinas/fisiología , Neointima/prevención & control , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo/fisiología , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Citocinas/genética , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Arteria Femoral/patología , Arteria Femoral/fisiopatología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/fisiología , Humanos , Lectinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Neointima/patología , Neointima/fisiopatología , Remodelación Vascular/fisiología
17.
FASEB J ; 29(7): 3085-99, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25854700

RESUMEN

Hyperhomocysteinemia (HHcy) impairs re-endothelialization and accelerates vascular remodeling. The role of CD34(+)/VEGF receptor (VEGFR) 2(+) progenitor cells (PCs) in vascular repair in HHcy is unknown. We studied the effect of HHcy on PCs and its role in vascular repair in severe HHcy (∼150 µM), which was induced in cystathionine-ß synthase heterozygous mice fed a high-methionine diet for 8 weeks. Vascular injury was introduced by carotid air-dry endothelium denudation. CD34(+)/VEGFR2(+) cells were examined by flow cytometry. HHcy reduced bone marrow (BM) CD34(+)/VEGFR2(+) cells and suppressed replenishment of postinjury CD34(+)/VEGFR2(+) cells in peripheral blood (PB). Donor green fluorescent protein-positive PC homing to the injured vessel was reduced in HHcy after CD34(+) PCs from enhanced green fluorescent protein mice were adoptively transferred following carotid injury. CD34(+) PC transfusion partially reversed HHcy-suppressed re-endothelialization and HHcy-induced neointimal formation. Furthermore, homocysteine (Hcy) inhibited proliferation, adhesion, and migration and suppressed ß1-integrin expression and activity in human CD34(+) endothelial colony-forming cells (ECFCs) isolated from PBs in a dose-dependent manner. A functional-activating ß1-integrin antibody rescued Hcy-suppressed adhesion and migration in CD34(+) ECFCs. In conclusion, HHcy reduces BM CD34(+)/VEGFR2(+) generation and suppresses CD34(+)/VEGFR2(+) cell mobilization and homing to the injured vessel via ß1-integrin inhibition, which partially contributes to impaired re-endothelialization and vascular remodeling.


Asunto(s)
Células de la Médula Ósea/patología , Células de la Médula Ósea/fisiología , Hiperhomocisteinemia/patología , Hiperhomocisteinemia/fisiopatología , Integrina beta1/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígenos CD34/metabolismo , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , Adhesión Celular , Movimiento Celular , Ensayo de Unidades Formadoras de Colonias , Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/fisiología , Humanos , Hiperhomocisteinemia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neointima/patología , Neointima/fisiopatología , Neointima/prevención & control , Remodelación Vascular
18.
Arterioscler Thromb Vasc Biol ; 35(10): 2145-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26293467

RESUMEN

OBJECTIVE: To investigate the functional role of the microRNA (miR)-15b/16 in vascular smooth muscle (SM) phenotypic modulation. APPROACH AND RESULTS: We found that miR-15b/16 is one of the most abundant mRs expressed in contractile vascular smooth muscle cells (VSMCs). However, when contractile VSMCs get converted to a synthetic phenotype, miR-15b/16 expression is significantly reduced. Knocking down endogenous miR-15b/16 in VSMCs attenuates SM-specific gene expression but promotes VSMC proliferation and migration. Conversely, overexpression of miR-15b/16 promotes SM contractile gene expression while attenuating VSMC migration and proliferation. Consistent with this, overexpression of miR-15b/16 in a rat carotid balloon injury model markedly attenuates injury-induced SM dedifferentiation and neointima formation. Mechanistically, we identified the potent oncoprotein yes-associated protein (YAP) as a downstream target of miR-15b/16 in VSMCs. Reporter assays validated that miR-15b/16 targets YAP's 3' untranslated region. Moreover, overexpression of miR-15b/16 significantly represses YAP expression, whereas conversely, depletion of endogenous miR-15b/16 results in upregulation of YAP expression. CONCLUSIONS: These results indicate that miR-15b/16 plays a critical role in SM phenotypic modulation at least partly through targeting YAP. Restoring expression of miR-15b/16 would be a potential therapeutic approach for treatment of proliferative vascular diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Músculo Liso Vascular/citología , Neointima/genética , Animales , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/fisiopatología , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Neointima/fisiopatología , Fenotipo , Ratas , Sensibilidad y Especificidad , Proteínas Señalizadoras YAP
19.
Pathobiology ; 83(1): 47-52, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890264

RESUMEN

OBJECTIVE: The potential contribution of vascular endothelial growth factor (VEGF) in neointima development has been evaluated in numerous animal studies. However, its role remains controversial. Moreover, little is known about neointima formation in humans. In this study we assessed the expression of VEGF-A and its receptors in the human neointima formed within vascular anastomosis. METHODS: The studied material comprised neointima samples harvested during secondary vascular operations from patients with chronic limb ischemia after aorto-/iliofemoral bypass grafting who developed vascular graft occlusion at 6-18 months after the initial surgical treatment. The control material consisted of segments of femoral arteries without visible macroscopic lesions collected from organ donors. The expression and content of VEGF-A, VEGFR-1 and VEGFR-2 were analyzed with PCR and ELISA methods, respectively. RESULTS: We observed a significantly increased expression of VEGF-A and VEGFR-2 mRNA in neointima compared to the normal aorta. A significantly higher protein content of VEGF-A and VEGFR-2 in neointima samples compared to the controls was also observed. No significant difference of VEGFR-1 content and VEGFR-1 mRNA expression was found in the studied material. CONCLUSION: These results indicate a possible involvement of the VEGF-A and VEGFR-2 system in the pathologic process of human neointima formation after vascular interventions.


Asunto(s)
Neointima/genética , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Expresión Génica , Oclusión de Injerto Vascular , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/citología , Neointima/fisiopatología , Neovascularización Patológica , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Circ J ; 80(9): 1916-21, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27357218

RESUMEN

BACKGROUND: No previous study has reported a comprehensive comparison of the chronic angioscopic findings after bare metal stent (BMS), and 1st- and 2nd-generation drug-eluting stents (DES). METHODS AND RESULTS: The Multicenter Study on Intra-Coronary Angioscopy after Stent (MICASA) is a multicenter registry of coronary angioscopy. A total of 264 stents were observed by coronary angioscopy 1 year after PCI. There were 15 BMS, 90 1st-generation DES, and 159 2nd-generation DES. Neointimal coverage (NC) of the stent was classified into 4 grades from 0 (no coverage) to 3 (complete coverage). Yellow color (YC) of plaque at the stented segment was graded from 0 (white) to 3 (bright yellow). Minimum (Min-) and Maximum (Max-) NC grade were significantly lower with 1st- and 2nd-generation DES than with BMS. Although the Max-NC grade was similar, the Min-NC grade was significantly higher for 2nd-generation DES than for 1st-generation DES. Both the YC grade and the incidence of thrombus with 2nd-generation DES were lower than with the 1st-generation DES and were comparable to BMS. Multivariate analysis showed that low-density lipoprotein, 1st-generation DES, and acute coronary syndrome were independent factors for yellow plaque (YG2 or 3), and that hypertension and 1st-generation DES were independent factors for the incidence of thrombus. CONCLUSIONS: Coronary angioscopy revealed more homogeneous coverage with white neointima and less thrombus after 2nd-generation DES as compared with 1st-generation DES. These findings may explain the favorable clinical outcomes observed for patients treated with 2nd-generation DES. (Circ J 2016; 80: 1916-1921).


Asunto(s)
Angioscopía , Stents Liberadores de Fármacos , Lipoproteínas LDL/sangre , Neointima , Sistema de Registros , Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/epidemiología , Síndrome Coronario Agudo/fisiopatología , Síndrome Coronario Agudo/cirugía , Anciano , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Neointima/sangre , Neointima/epidemiología , Neointima/patología , Neointima/fisiopatología , Trombosis/sangre , Trombosis/etiología , Trombosis/patología , Trombosis/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA