Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Biomed Pharmacother ; 143: 112165, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34543986

RESUMEN

The injury of endothelial cells is one of the initiating factors in restenosis after endovascular treatment. Human urinary kallidinogenase (HUK) is a tissue kallikrein which is used for ischemia-reperfusion injury treatment. Studies have shown that HUK may be a potential therapeutic agent to prevent stenosis after vascular injury, however, the precise mechanisms have not been fully established. This study is to investigate whether HUK can protect endothelial cells after balloon injury or H2O2-induced endothelial cell damage through the proline-rich tyrosine kinase 2 (Pyk2)/mitochondrial calcium uniporter (MCU) pathway. Intimal hyperplasia, a decrease of pinocytotic vesicles and cell apoptosis were found in the common carotid artery balloon injury and H2O2-induced endothelial cell damage, Pyk2/MCU was also up-regulated in such pathological process. HUK could prevent these injuries partially via the bradykinin B2 receptor by inhibiting Pyk2/MCU pathway, which prevented the mitochondrial damage, maintained calcium balance, and eventually inhibited cell apoptosis. Furthermore, MCU expression was not markedly increased if Pyk2 was suppressed by shRNA technique in the H2O2 treatment group, and cell viability was significantly better than H2O2-treated only. In short, our results indicate that the Pyk2/MCU pathway is involved in endothelial injury induced by balloon injury or H2O2-induced endothelial cell damage. HUK plays an protective role by inhibiting the Pyk2/MCU pathway in the endothelial injury.


Asunto(s)
Canales de Calcio/metabolismo , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Arteria Carótida Común/efectos de los fármacos , Quinasa 2 de Adhesión Focal/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Calicreínas/farmacología , Animales , Apoptosis/efectos de los fármacos , Canales de Calcio/genética , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/ultraestructura , Células Cultivadas , Modelos Animales de Enfermedad , Quinasa 2 de Adhesión Focal/genética , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Peróxido de Hidrógeno/toxicidad , Calicreínas/orina , Masculino , Neointima , Ratas Sprague-Dawley , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal
2.
J Vasc Res ; 57(6): 325-340, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32777783

RESUMEN

We have shown that both insulin and resveratrol (RSV) decrease neointimal hyperplasia in chow-fed rodents via mechanisms that are in part overlapping and involve the activation of endothelial nitric oxide synthase (eNOS). However, this vasculoprotective effect of insulin is abolished in high-fat-fed insulin-resistant rats. Since RSV, in addition to increasing insulin sensitivity, can activate eNOS via pathways that are independent of insulin signaling, such as the activation of sirtuin 1 (SIRT1) and AMP-activated kinase (AMPK), we speculated that unlike insulin, the vasculoprotective effect of RSV would be retained in high-fat-fed rats. We found that high-fat feeding decreased insulin sensitivity and increased neointimal area and that RSV improved insulin sensitivity (p < 0.05) and decreased neointimal area in high-fat-fed rats (p < 0.05). We investigated the role of SIRT1 in the effect of RSV using two genetic mouse models. We found that RSV decreased neointimal area in high-fat-fed wild-type mice (p < 0.05), an effect that was retained in mice with catalytically inactive SIRT1 (p < 0.05) and in heterozygous SIRT1-null mice. In contrast, the effect of RSV was abolished in AMKPα2-null mice. Thus, RSV decreased neointimal hyperplasia after arterial injury in both high-fat-fed rats and mice, an effect likely not mediated by SIRT1 but by AMPKα2.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Arteria Carótida Común/efectos de los fármacos , Dieta Alta en Grasa , Arteria Femoral/efectos de los fármacos , Neointima , Resveratrol/farmacología , Sirtuina 1/metabolismo , Lesiones del Sistema Vascular/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/genética , Animales , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Modelos Animales de Enfermedad , Arteria Femoral/enzimología , Arteria Femoral/lesiones , Arteria Femoral/patología , Resistencia a la Insulina , Ratones Noqueados , Ratas Sprague-Dawley , Transducción de Señal , Sirtuina 1/genética , Lesiones del Sistema Vascular/enzimología , Lesiones del Sistema Vascular/patología
3.
Microvasc Res ; 132: 104060, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32818511

RESUMEN

BACKGROUND: Cardiovascular diseases are associated with proliferation and phenotypic switch. Platelet-derived growth factor-BB (PDGF-BB) is a major initiating factor for proliferative vascular diseases, such as neointimal lesion formation, restenosis after angioplasty, and atherosclerosis. Ruxolitinib, a potent Janus kinase (JAK) 1 and 2 inhibitor, has been reported to significantly block the proliferation-related signaling pathway of JAK2/signal transducers and activators of transcription 3 (STAT3) and harbor a broad spectrum of anti-cancer activities, including proliferation inhibition, apoptosis induction, and anti-inflammation. However, the role of ruxolitinib in regulating PDGF-BB-induced VSMC proliferation remains to be elucidated. Thus, this study investigates the role of ruxolitinib in regulating PDGF-BB-induced VSMC proliferation and its underlying mechanisms. METHODS: In vivo, the medial thickness of the carotid artery was evaluated using a mouse carotid ligation model, ruxolitinib was administered orally to the mice every other day, and the mice were euthanized on day 28 to evaluate the therapeutic effects of ruxolitinib. Cell proliferation markers were measured using real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting. In vitro, VSMCs were treated with ruxolitinib with or without PDGF-BB at an indicated time and concentration. Cell proliferation and apoptosis were measured using Cell Counting Kit-8 assay, MTS assays and flow cytometry. The JAK2/STAT3 signaling pathway involved in the effects of ruxolitinib on VSMCs was detected by western blotting with the specific pathway inhibitor AG490. RESULTS: In vivo, ruxolitinib significantly decreased the ratio-of-intima ratio (I/M ratio) by inhibiting the expression of PCNA and cyclinD1 (p <0.05). In vitro, ruxolitinib inhibited PDGF-BB-induced VSMC proliferation compared with the PDGF-BB treatment group (p <0.05). In addition, ruxolitinib inhibited the PDGF-BB-induced activation of the JAK2/STAT3 signaling pathway and decreased the expression of proliferation related-proteins cyclinD1 and PCNA in VSMCs (p <0.05). CONCLUSION: Our findings suggest that ruxolitinib inhibits VSMC proliferation in vivo and in vitro by suppressing the activation of the JAK2/STAT3 signaling pathway. Therefore, ruxolitinib has a therapeutic potential for proliferative vascular diseases.


Asunto(s)
Becaplermina/farmacología , Estenosis Carotídea/prevención & control , Janus Quinasa 2/metabolismo , Inhibidores de las Cinasas Janus/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Neointima , Pirazoles/farmacología , Factor de Transcripción STAT3/metabolismo , Animales , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Estenosis Carotídea/enzimología , Estenosis Carotídea/patología , Células Cultivadas , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Hiperplasia , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Nitrilos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Pirimidinas , Transducción de Señal
4.
Arterioscler Thromb Vasc Biol ; 40(7): 1705-1721, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268790

RESUMEN

OBJECTIVE: A decrease in nitric oxide, leading to vascular smooth muscle cell proliferation, is a common pathological feature of vascular proliferative diseases. Nitric oxide synthesis by eNOS (endothelial nitric oxide synthase) is precisely regulated by protein kinases including AKT1. ENH (enigma homolog protein) is a scaffolding protein for multiple protein kinases, but whether it regulates eNOS activation and vascular remodeling remains unknown. Approach and Results: ENH was upregulated in injured mouse arteries and human atherosclerotic plaques and was associated with coronary artery disease. Neointima formation in carotid arteries, induced by ligation or wire injury, was greatly decreased in endothelium-specific ENH-knockout mice. Vascular ligation reduced AKT and eNOS phosphorylation and nitric oxide production in the endothelium of control but not ENH-knockout mice. ENH was found to interact with AKT1 and its phosphatase PHLPP2 (pleckstrin homology domain and leucine-rich repeat protein phosphatase 2). AKT and eNOS activation were prolonged in VEGF (vascular endothelial growth factor)-induced ENH- or PHLPP2-deficient endothelial cells. Inhibitors of either AKT or eNOS effectively restored ligation-induced neointima formation in ENH-knockout mice. Moreover, endothelium-specific PHLPP2-knockout mice displayed reduced ligation-induced neointima formation. Finally, PHLPP2 was increased in the endothelia of human atherosclerotic plaques and blood cells from patients with coronary artery disease. CONCLUSIONS: ENH forms a complex with AKT1 and its phosphatase PHLPP2 to negatively regulate AKT1 activation in the artery endothelium. AKT1 deactivation, a decrease in nitric oxide generation, and subsequent neointima formation induced by vascular injury are mediated by ENH and PHLPP2. ENH and PHLPP2 are thus new proatherosclerotic factors that could be therapeutically targeted.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Traumatismos de las Arterias Carótidas/enzimología , Arteria Carótida Común/enzimología , Proteínas de Microfilamentos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Remodelación Vascular , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Aterosclerosis/enzimología , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , Arteria Carótida Común/patología , Arteria Carótida Común/fisiopatología , Células Cultivadas , Enfermedad de la Arteria Coronaria/enzimología , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/fisiopatología , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Neointima , Óxido Nítrico/metabolismo , Fosfoproteínas Fosfatasas/deficiencia , Fosfoproteínas Fosfatasas/genética , Fosforilación , Transducción de Señal
5.
Atherosclerosis ; 299: 15-23, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32182440

RESUMEN

BACKGROUND AND AIMS: Exercise training (ET) helps treat atherosclerosis. However, many patients stop regular ET for various reasons. The effect of detraining on atherosclerosis is not well studied. We examined the effects of ET vs. short-term detraining on atheromatous matrix-metalloproteinase (MMP) activity in preexisting plaque and circulating cytokines/lipids. METHODS AND RESULTS: Eighteen-week-old apolipoprotein-E-/- mice (n = 56) on a Western diet underwent: 1) ET for 6-weeks (ET5+1), 2) ET for 5-weeks and detraining for 1-week (ET5+0), 3) ET for the last 1-week (ET0+1), or 4) no treadmill ET at all for 6-weeks (ET0+0). Atheromatous MMP-activity was visualized using molecular imaging with an MMP-2/9-activatable near-infrared-fluorescent probe. Compared with no ET (ET0+0), regular ET (ET5+1) decreased carotid atheromatous MMP activity, but this protective effect was significantly blunted by short-term detraining (ET5+0). Short-term detraining after longer-term ET showed a reduction in MMP-activity similar to short-term ET (ET0+1). Blood levels of lipids and cytokines paralleled the molecular imaging results: exercise caused higher levels of high-density lipoprotein, adiponectin, and interleukin-10 and lower levels of vascular cell adhesion molecule, monocyte chemoattractant protein-1, interleukin-1ß, and low-density lipoprotein. However, this beneficial effect was short-lived, with the ET5+0 group being similar to the ET0+0 group, and the ET0+1 group being similar to the ET5+1 group. The effect of exercise can be modeled with an exponential-decay of the protective factor of about 15%/day. CONCLUSIONS: Even short-term detraining reduces atheroprotective effects, and tips the balance towards atherosclerosis. This suggests that ET, to be effective, needs to be prolonged and regular, and that detraining should be avoided.


Asunto(s)
Enfermedades de las Arterias Carótidas/terapia , Arteria Carótida Común/enzimología , Terapia por Ejercicio , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Conducta Sedentaria , Animales , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/enzimología , Enfermedades de las Arterias Carótidas/patología , Arteria Carótida Común/patología , Citocinas/sangre , Modelos Animales de Enfermedad , Lípidos/sangre , Ratones Noqueados para ApoE , Placa Aterosclerótica , Carrera , Factores de Tiempo
6.
Heart Vessels ; 35(2): 278-283, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31501952

RESUMEN

COX-2 contributes to local inflammation in atherosclerotic lesions. Regulatory T cells (Tregs) enhance the stability of atherosclerotic plaques. The aim of this study was to detect the potential relationship between Tregs and COX-2 in vulnerable plaques. Thirty ApoE -/- mice were fed a high-fat diet, and a silastic perivascular collar was placed around the right common carotid artery to induce vulnerable plaques. Eight weeks after collar placement, the mice were divided randomly into three groups: control, PBS, and Treg groups. Four weeks later, the right common carotid arteries were collected to detect the expression of COX-2. The results showed that Tregs significantly suppressed the expression of COX-2 in vulnerable plaques. In an in vitro experiment, RAW264.7 cells were divided randomly into three groups, which were precultured without T cells or with CD4 + CD25- T cells or Tregs for 48 h with an anti-CD3 antibody; then the cells were stimulated with LPS for 24 h. The RAW264.7 cells were harvested for RT-PCR and western blot assays and the results showed that Tregs downregulated COX-2 expression in RAW264.7 cells. Therefore, Tregs inhibited the expression of COX-2 in vulnerable plaques and macrophages, and COX-2 inhibition may be an important effect of Tregs that results in atherosclerotic plaque stabilization.


Asunto(s)
Enfermedades de las Arterias Carótidas/enzimología , Arteria Carótida Común/enzimología , Comunicación Celular , Ciclooxigenasa 2/metabolismo , Placa Aterosclerótica , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/prevención & control , Arteria Carótida Común/inmunología , Arteria Carótida Común/patología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Células RAW 264.7 , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante
7.
Am J Physiol Heart Circ Physiol ; 317(5): H969-H980, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31518169

RESUMEN

Multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a multigene family with isoform-specific regulation of vascular smooth muscle (VSM) functions. In previous studies, we found that vascular injury resulted in VSM dedifferentiation and reduced expression of the CaMKIIγ isoform in medial wall VSM. Smooth muscle knockout of CaMKIIγ enhanced injury-induced VSM neointimal hyperplasia, whereas CaMKIIγ overexpression inhibited VSM proliferation and neointimal formation. In this study, we evaluated DNA cytosine methylation/demethylation as a mechanism for regulating CaMKII isoform expression in VSM. Inhibition of cytosine methylation with 5-Aza-2'-deoxycytidine significantly upregulated CaMKIIγ expression in cultured VSM cells and inhibited CaMKIIγ downregulation in organ-cultured aorta ex vivo. With the use of methylated cytosine immunoprecipitation, the rat Camk2g promoter was found hypomethylated in differentiated VSM, whereas injury- or cell culture-induced VSM dedifferentiation coincided with Camk2g promoter methylation and decreased expression. We report for the first time that VSM cell phenotype switching is accompanied by marked induction of thymine DNA glycosylase (TDG) protein and mRNA expression in injured arteries in vivo and in cultured VSM synthetic phenotype cells. Silencing Tdg in VSM promoted expression of CaMKIIγ and differentiation markers, including myocardin, and inhibited VSM cell proliferation and injury-induced neointima formation. This study indicates that CaMKIIγ expression in VSM is regulated by cytosine methylation/demethylation and that TDG is an important determinant of this process and, more broadly, VSM phenotype switching and function.NEW & NOTEWORTHY Expression of the calcium calmodulin-dependent protein kinase II-γ isoform (CaMKIIγ) is associated with differentiated vascular smooth muscle (VSM) and negatively regulates proliferation in VSM synthetic phenotype (VSMSyn) cells. This study demonstrates that thymine DNA glycosylase (TDG) plays a key role in regulating CaMKIIγ expression in VSM through promoter cytosine methylation/demethylation. TDG expression is strongly induced in VSMSyn cells and plays key roles in negatively regulating CaMKIIγ expression and more broadly VSM phenotype switching.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Traumatismos de las Arterias Carótidas/enzimología , Plasticidad de la Célula , Metilación de ADN , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Timina ADN Glicosilasa/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Neointima , Fenotipo , Regiones Promotoras Genéticas , Ratas Sprague-Dawley , Transducción de Señal , Timina ADN Glicosilasa/genética
8.
Ann Vasc Surg ; 57: 201-209, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30684618

RESUMEN

BACKGROUND: Intimal hyperplasia (IH) is the most common indicator for secondary intervention in peripheral vascular disease. Matrix metalloproteinases (MMPs) play a role in IH development due to their degradation of the extracellular matrix. Doxycycline (Doxy), a member of the tetracycline family of antibiotics, is a potent MMP inhibitor. We have previously shown that Doxy inhibits MMP activity and vascular smooth muscle cell migration in vitro. We hypothesized that Doxy would decrease MMP activity in vivo and inhibit the development of IH in a rodent model of vascular injury. METHODS AND RESULTS: Doxy (400 mg/pellet) was delivered by a slow-release pellet implanted 3 days prior to or at the time of balloon angioplasty (BA) of the common carotid artery in female rats. At 14 days post-BA, intima-to-media (I:M) ratios were 0.77 ± 0.21 and 1.04 ± 0.32 in the Doxy treated groups, respectively, compared to 1.25 ± 0.26 in the control group (P = not significant; n = 3). Additionally, the tested dose of Doxy in either group had no inhibitory effect on membrane type 1-MMP or MMP-2 tissue levels, as measured by immunohistochemistry, or on systemic levels of MMP, as measured by total MMP serum levels using enzyme-linked immunosorbent assay. At 14 days post-BA, VSMC proliferation in the injured artery was increased to Doxy treatment prior to and at the time of surgery (23.5 ± 3.4 and 27.2 ± 3.9%, respectively), compared to control (11.4 ± 0.4%; n = 3), as measured by proliferating cellular nuclear antigen immunostaining. CONCLUSIONS: In our in vivo model of vascular injury, systemic Doxy administration prior to or at the time of vascular injury does not significantly hinder the progression of IH development. Additional doses and routes of administration could be examined in order to correlate therapeutic serum levels of Doxy with effective MMP inhibition in serum and arterial tissue. However, alternative drug delivery systems are needed in order to optimize therapeutic administration of targeted MMP inhibitors for the prevention of IH development.


Asunto(s)
Angioplastia de Balón/efectos adversos , Fármacos Cardiovasculares/administración & dosificación , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Doxiciclina/administración & dosificación , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Neointima , Animales , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Hiperplasia , Metaloproteinasa 14 de la Matriz/sangre , Metaloproteinasa 2 de la Matriz/sangre , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Ratas Sprague-Dawley
9.
Biomed Pharmacother ; 106: 1091-1097, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30119175

RESUMEN

Ginsenoside Re (GS-Re) is one of the main ingredients of ginseng, a widely known Chinese traditional medicine, and has a variety of beneficial effects, including vasorelaxation, antioxidative, anti-inflammatory, and anticancer properties. The aims of the present study were to observe the effect of GS-Re on balloon injury-induced neointimal hyperplasia in the arteries and to investigate the mechanisms underlying this effect. A rat vascular neointimal hyperplasia model was generated by rubbing the endothelium of the common carotid artery (CCA) with a balloon, and GS-Re (12.5, 25 or 50 mg/kg/d) were subsequently continuously administered to the rats by gavage for 14 days. After GS-Re treatment, the vessel lumen of injured vessels showed significant increases in the GS-Re 25.0 and 50.0 mg/kg/d (intermediate- and high-dose) groups according to H.E. staining. Additionally, a reduced percentage of proliferating cell nuclear antigen (PCNA)-positive cells and an increased number of SM α-actin-positive cells were detected, and the levels of NO, cyclic guanosine monophosphate (cGMP), and eNOS mRNA as well as the phos-eNOSser1177/eNOS protein ratio were obviously upregulated in the intermediate- and high-dose groups. Moreover, the promotive effects of GS-Re on NO and eNOS expression were blocked by L-NAME treatment to different degrees. These results suggested that GS-Re can suppress balloon injury-induced vascular neointimal hyperplasia by inhibiting VSMC proliferation, which is closely related to the activation of the eNOS/NO/cGMP pathway.


Asunto(s)
Angioplastia de Balón/instrumentación , Traumatismos de las Arterias Carótidas/prevención & control , Arteria Carótida Común/efectos de los fármacos , GMP Cíclico/metabolismo , Ginsenósidos/farmacología , Neointima , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Actinas/metabolismo , Animales , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hiperplasia , Masculino , Óxido Nítrico Sintasa de Tipo III/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas Sprague-Dawley , Sistemas de Mensajero Secundario/efectos de los fármacos
10.
Atherosclerosis ; 266: 212-222, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29053988

RESUMEN

BACKGROUND AND AIMS: Vascular smooth muscle cell (VSMC) dedifferentiation contributes to neointima formation, which results in various vascular disorders. Intermedin (IMD), a cardiovascular paracrine/autocrine polypeptide, is involved in maintaining circulatory homeostasis. However, whether IMD protects against neointima formation remains largely unknown. The purpose of this study is to investigate the role of IMD in neointima formation and the possible mechanism. METHODS: IMD1-53 (100ng/kg/h) or saline water was used on rat carotid-artery balloon-injury model. The mouse left common carotid-artery ligation-injury model was established using IMD-transgenic and C57BL/6J mice. Immunohistochemistry and immunofluorescence staining was used to detect the protein expression in rat carotid arteries. Radioimmunoassay was used to determine the serum IMD level. The hematoxylin andeosin staining was used for carotid arteries morphological testing. In vitro, for rat primary cultured VSMC phenotype transition, proliferation and migration assays, platelet-derived growth factor-BB (PDGF-BB) reagent and IMD1-53 peptide were added to the culture media at the final concentration of 20 ng/mL and 10-7mol/L respectively. Quantification of VSMC proliferation involved MTT and BrdU assay and migration was detected by wound-healing assay. Western blot and realtime PCR were used to detect the protein and mRNA levels of tissues or cells. RESULTS: With the rat carotid-artery balloon-injury model, IMD was significantly downregulated in injured arteries and plasma. Exogenous IMD1-53 greatly inhibited neointima formation and prevented VSMC from switching to a synthetic phenotype. With the left common carotid-artery ligation-injury model, IMD-transgenic mice showed less neointima formation than C57BL/6J mice. PDGF-BB reduced IMD mRNA expression in rat primary cultured VSMCs but increased that of its receptors, calcitonin receptor-like receptor or receptor activity-modifying proteins. Furthermore, PDGF-BB promoted VSMC proliferation and migration and transformed VSMCs to the synthetic phenotype, which was reversed with IMD1-53 treatment. Mechanistically, IMD1-53 maintained the contractile VSMC phenotype via the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) pathway. CONCLUSIONS: IMD attenuated neointima formation both in the rat model of carotid-artery balloon injury and mouse model of common carotid-artery ligation injury. IMD protection may be mediated by maintaining a VSMC contractile phenotype via the cAMP/PKA pathway.


Asunto(s)
Adrenomedulina/metabolismo , Traumatismos de las Arterias Carótidas/enzimología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Neointima , Neuropéptidos/metabolismo , Adrenomedulina/genética , Animales , Becaplermina , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Movimiento Celular , Proliferación Celular , Transdiferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Neuropéptidos/genética , Fenotipo , Proteínas Proto-Oncogénicas c-sis/farmacología , Ratas Sprague-Dawley , Sistemas de Mensajero Secundario , Factores de Tiempo , Vasoconstricción
11.
J Vasc Res ; 54(3): 180-192, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28490016

RESUMEN

BACKGROUND: Pituitary adenylate cyclase-activating polypeptide (PACAP) is a multifunctional neuropeptide in the VIP/secretin/glucagon peptide superfamily. Two active forms, PACAP1-38 and PACAP1-27, act through G protein-coupled receptors, the PAC1 and VPAC1/2 receptors. Effects of PACAP include potent vasomotor activity. Vasomotor activity and organ-specific vasomotor effects of PACAP-deficient mice have not yet been investigated; thus, the assessment of its physiological importance in vasomotor functions is still missing. We hypothesized that backup mechanisms exist to maintain PACAP pathway activity in PACAP knockout (KO) mice. Thus, we investigated the vasomotor effects of exogenous vasoactive intestinal peptide (VIP) and PACAP polypeptides in PACAP wild-type (WT) and PACAP-deficient (KO) male mice. METHODS: Carotid and femoral arteries were isolated from 8- to 12-week-old male WT and PACAP-KO mice. Vasomotor responses were measured with isometric myography. RESULTS: In the arteries of WT mice the peptides induced relaxations, which were significantly greater to PACAP1-38 than to PACAP1-27 and VIP. In KO mice, PACAP1-38 did not elicit relaxation, whereas PACAP1-27 and VIP elicited significantly greater relaxation in KO mice than in WT mice. The specific PAC1R and VPAC1R antagonist completely blocked the PACAP-induced relaxations. CONCLUSION: Our data suggest that in PACAP deficiency, backup mechanisms maintain arterial relaxations to polypeptides, indicating an important physiological role for the PACAP pathway in the regulation of vascular tone.


Asunto(s)
Arteria Carótida Común/efectos de los fármacos , Arteria Femoral/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/deficiencia , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Péptido Intestinal Vasoactivo/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Arteria Carótida Común/enzimología , Relación Dosis-Respuesta a Droga , Arteria Femoral/enzimología , Genotipo , Técnicas In Vitro , Masculino , Ratones Noqueados , Fenotipo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/agonistas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Nucl Med Commun ; 38(4): 299-305, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28234785

RESUMEN

OBJECTIVE: Previous studies have suggested that matrix metalloproteinase (MMP) inhibitor uptake may offer a precise estimation of MMP activity in atherosclerotic lesions. In this study, we explored the feasibility of noninvasive detection of MMP-9 activity using technetium-99m-labeled matrix metalloproteinase-9 antibody (Tc-McAb) in vivo. METHODS: ApoE-deficient (ApoE) atherosclerosis mice models (n=10) were induced through a high-cholesterol diet following ligation of their left common carotid artery. After 4 weeks, the models were verified through proton density-weighted and T2-weighted images obtained by MRI. C57BL/6 sham mice (n=8) were used as controls. In addition, normal mice (n=20) were used to characterize blood clearance. After radiolabeled McAb administration, single-photon emission computed tomography (SPECT) was performed. Subsequently, left common carotid arteries were harvested for ex-vivo autoradiograph imaging. Then, morphology and activity assays of MMP-9 were histologically and immunohistochemically examined. RESULTS: MRI showed higher signal intensities in the left common carotid arteries with irregular stenoses in the lumen of blood vessels in atherosclerosis mice models in vivo. Atherosclerotic lesions on left common carotid artery specimens were also clearly visualized using SPECT 2 h after Tc-McAb administration in vivo. Note that the radiochemistry purity of the Tc-McAb used was 85-95%. Biodistribution studies have shown that the clearance of Tc-McAb from blood was rapid. In addition, atherosclerotic lesions were clearly visualized on radioautography film shadows ex vivo. CONCLUSION: MMP-9 activities within the atherosclerotic lesions were noninvasively detected using Tc-labeled SPECT in vivo.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Aterosclerosis/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Anticuerpos Monoclonales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Autorradiografía , Arteria Carótida Común/enzimología , Modelos Animales de Enfermedad , Estudios de Factibilidad , Inmunohistoquímica , Masculino , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Radiofármacos , Tecnecio
13.
Arterioscler Thromb Vasc Biol ; 37(3): 476-484, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28062495

RESUMEN

OBJECTIVE: RNA polymerase I (Pol I)-dependent rRNA synthesis is a determinant factor in ribosome biogenesis and thus cell proliferation. The importance of dysregulated Pol I activity in cardiovascular disease, however, has not been recognized. Here, we tested the hypothesis that specific inhibition of Pol I might prevent arterial injury-induced neointimal hyperplasia. APPROACH AND RESULTS: CX-5461 is a novel selective Pol I inhibitor. Using this tool, we demonstrated that local inhibition of Pol I blocked balloon injury-induced neointima formation in rat carotid arteries in vivo. Neointimal development was associated with augmented rDNA transcriptional activity as evidenced by the increased phosphorylation of upstream binding factor-1. The beneficial effect of CX-5461 was mainly mediated by inducing G2/M cell cycle arrest of proliferating smooth muscle cells without obvious apoptosis. CX-5461 did not induce p53 stabilization but increased p53 phosphorylation and acetylation and activated the ataxia telangiectasia mutated/ataxia telangiectasia and Rad3-related (ATR) pathway. Inhibition of ATR, but not of ataxia telangiectasia mutated, abolished the cytostatic effect of CX-5461 and p53 phosphorylation. In addition, inhibition of p53 or knockdown of the p53 target GADD45 mimicked the effect of ATR inhibition. In vivo experiments showed that the levels of phospho-p53 and acetyl-p53, and activity of the ataxia telangiectasia mutated/ATR pathway were all augmented in CX-5461-treated vessels. CONCLUSIONS: Pol I can be therapeutically targeted to inhibit the growth of neointima, supporting that Pol I is a novel biological target for preventing arterial restenosis. Mechanistically, Pol I inhibition elicited G2/M cell cycle arrest in smooth muscle cells via activation of the ATR-p53 axis.


Asunto(s)
Benzotiazoles/farmacología , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Músculo Liso Vascular/efectos de los fármacos , Naftiridinas/farmacología , Neointima , ARN Polimerasa I/antagonistas & inhibidores , Acetilación , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Hiperplasia , Masculino , Ratones , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Fosforilación , ARN Polimerasa I/metabolismo , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
14.
J Cell Physiol ; 232(11): 3088-3102, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28019664

RESUMEN

Modulation from contractile to synthetic phenotype of vascular smooth muscle cells is a central process in disorders involving compromised integrity of the vascular wall. Phenotype modulation has been shown to include transition from voltage-dependent toward voltage-independent regulation of the intracellular calcium level, and inhibition of non-voltage dependent calcium influx contributes to maintenance of the contractile phenotype. One possible mediator of calcium-dependent signaling is the FAK-family non-receptor protein kinase Pyk2, which is activated by a number of stimuli in a calcium-dependent manner. We used the Pyk2 inhibitor PF-4594755 and Pyk2 siRNA to investigate the role of Pyk2 in phenotype modulation in rat carotid artery smooth muscle cells and in cultured intact arteries. Pyk2 inhibition promoted the expression of smooth muscle markers at the mRNA and protein levels under stimulation by FBS or PDGF-BB and counteracted phenotype shift in cultured intact carotid arteries and balloon injury ex vivo. During long-term (24-96 hr) treatment with PF-4594755, smooth muscle markers increased before cell proliferation was inhibited, correlating with decreased KLF4 expression and differing from effects of MEK inhibition. The Pyk2 inhibitor reduced Orai1 and preserved SERCA2a expression in carotid artery segments in organ culture, and eliminated the inhibitory effect of PDGF stimulation on L-type calcium channel and large-conductance calcium-activated potassium channel expression in carotid cells. Basal intracellular calcium level, calcium wave activity, and store-operated calcium influx were reduced after Pyk2 inhibition of growth-stimulated cells. Pyk2 inhibition may provide an interesting approach for preserving vascular smooth muscle differentiation under pathophysiological conditions.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Diferenciación Celular/efectos de los fármacos , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Vasoconstricción/efectos de los fármacos , Animales , Becaplermina , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/efectos de los fármacos , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/fisiopatología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/enzimología , Arteria Carótida Común/fisiopatología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasa 2 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/enzimología , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Técnicas de Cultivo de Órganos , Fenotipo , Proteínas Proto-Oncogénicas c-sis/farmacología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores de Tiempo , Transfección
15.
Arterioscler Thromb Vasc Biol ; 36(8): 1549-57, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27365406

RESUMEN

OBJECTIVE: Cathepsin S (CatS) participates in atherogenesis through several putative mechanisms. The ability of cathepsins to modify histone tail is likely to contribute to stem cell development. Histone deacetylase 6 (HDAC6) is required in modulating the proliferation and migration of various types of cancer cells. Here, we investigated the cross talk between CatS and HADC6 in injury-related vascular repair in mice. APPROACH AND RESULTS: Ligation injury to the carotid artery in mice increased the CatS expression, and CatS-deficient mice showed reduced neointimal formation in injured arteries. CatS deficiency decreased the phosphorylation levels of p38 mitogen-activated protein kinase, Akt, and HDAC6 and toll-like receptor 2 expression in ligated arteries. The genetic or pharmacological inhibition of CatS also alleviated the increased phosphorylation of p38 mitogen-activated protein kinase, Akt, and HDAC6 induced by platelet-derived growth factor BB in cultured vascular smooth muscle cells (VSMCs), and p38 mitogen-activated protein kinase inhibition and Akt inhibition decreased the phospho-HDAC6 levels. Moreover, CatS inhibition caused decrease in the levels of the HDAC6 activity in VSMCs in response to platelet-derived growth factor BB. The HDAC6 inhibitor tubastatin A downregulated platelet-derived growth factor-induced VSMC proliferation and migration, whereas HDAC6 overexpression exerted the opposite effect. Tubastatin A also decreased the intimal VSMC proliferation and neointimal hyperplasia in response to injury. Toll-like receptor 2 silencing decreased the phosphorylation levels of p38 mitogen-activated protein kinase, Akt, and HDAC6 and VSMC migration and proliferation. CONCLUSIONS: This is the first report detailing cross-interaction between toll-like receptor 2-mediated CatS and HDAC6 during injury-related vascular repair. These data suggest that CatS/HDAC6 could be a potential therapeutic target for the control of vascular diseases that are involved in neointimal lesion formation.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Arteria Carótida Común/enzimología , Catepsinas/metabolismo , Histona Desacetilasas/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Toll-Like 2/metabolismo , Cicatrización de Heridas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/patología , Catepsinas/antagonistas & inhibidores , Catepsinas/deficiencia , Catepsinas/genética , Puntos de Control del Ciclo Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Genotipo , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Masculino , Ratones Noqueados , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Neointima , Fenotipo , Fosforilación , Inhibidores de Proteasas/farmacología , Interferencia de ARN , Transducción de Señal , Receptor Toll-Like 2/genética , Transfección , Remodelación Vascular , Cicatrización de Heridas/efectos de los fármacos
16.
J Cardiovasc Transl Res ; 9(2): 135-44, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26928596

RESUMEN

To determine whether caspase-1 is critical in chronic kidney disease (CKD)-mediated arterial neointimal hyperplasia (NH), we utilized caspase(-/-) mice and induced NH in carotid artery in a CKD environment, and uremic sera-stimulated human vascular smooth muscle cells (VSMC). We made the following findings: (1) Caspase-1 inhibition corrected uremic sera-mediated downregulation of VSMC contractile markers, (2) CKD-promoted NH was attenuated in caspase(-/-) mice, (3) CKD-mediated downregulation of contractile markers was rescued in caspase null mice, and (4) expression of VSMC migration molecule αvß3 integrin was reduced in caspase(-/-) tissues. Our results suggested that caspase-1 pathway senses CKD metabolic danger signals. Further, CKD-mediated increase of contractile markers in VSMC and increased expression of VSMC migration molecule αvß3 integrin in NH formation were caspase-1 dependent. Therefore, caspase-1 is a novel therapeutic target for the suppression of CKD-promoted NH.


Asunto(s)
Enfermedades de las Arterias Carótidas/enzimología , Caspasa 1/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Neointima , Insuficiencia Renal Crónica/enzimología , Animales , Biomarcadores/metabolismo , Nitrógeno de la Urea Sanguínea , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/prevención & control , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Arteria Carótida Común/fisiopatología , Caspasa 1/deficiencia , Caspasa 1/genética , Inhibidores de Caspasas/farmacología , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Genotipo , Humanos , Hiperplasia , Integrina alfaVbeta3/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fenotipo , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/genética
17.
Arterioscler Thromb Vasc Biol ; 36(2): e9-16, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26586662

RESUMEN

OBJECTIVE: Hyaluronan (HA) is a polymeric glucosaminoglycan that forms a provisional extracellular matrix in diseased vessels. HA is synthesized by 3 different HA synthases (HAS1, HAS2, and HAS3). Aim of this study was to unravel the role of the HAS3 isoenzyme during experimental neointimal hyperplasia. APPROACH AND RESULTS: Neointimal hyperplasia was induced in Has3-deficient mice by ligation of the carotid artery. HA in the media of Has3-deficient mice was decreased 28 days after ligation, and neointimal hyperplasia was strongly inhibited. However, medial and luminal areas were unaffected. Cell density, proliferation, and apoptosis were not altered, suggesting a proportional decrease of both, the number of cells and extracellular matrix. In addition, endothelial function as determined by acetylcholine-induced relaxation of aortic rings, immunoblotting of endothelial nitric oxide synthase, and arterial blood pressure were not affected. Furthermore, the oxidative stress response was not affected as determined in total protein extracts from aortae. Transcriptome analysis comparing control versus ligated carotid arteries hinted toward a mitigated differential regulation of various signaling pathways in Has3-deficient mice in response to ligation that were related to vascular smooth muscle cell (VSMC) migration, including focal adhesions, integrins, mitogen-activated protein kinase, and phosphatidylinositol signaling system. Lentiviral overexpression of HAS3 in VSMC supported the migratory phenotype of VSMC in response to platelet-derived growth factor BB in vitro. Accordingly, knockdown of HAS3 reduced the migratory response to platelet-derived growth factor BB and in addition decreased the expression of PDGF-B mRNA. CONCLUSIONS: HAS3-mediated HA synthesis after vessel injury supports seminal signaling pathways in activation of VSMC, increases platelet-derived growth factor BB-mediated migration, and in turn enhances neointimal hyperplasia in vivo.


Asunto(s)
Enfermedades de las Arterias Carótidas/enzimología , Glucuronosiltransferasa/deficiencia , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Neointima , Animales , Becaplermina , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Genotipo , Glucuronosiltransferasa/genética , Hialuronano Sintasas , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fenotipo , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Transducción de Señal , Transcripción Genética , Transfección
18.
Arterioscler Thromb Vasc Biol ; 35(8): 1798-804, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26069236

RESUMEN

OBJECTIVE: Clinical evidence suggests an association between oxidative stress and vascular disease, and in vitro studies have demonstrated that reactive oxygen species can have prothrombotic effects on vascular and blood cells. It remains unclear, however, whether elevated levels of reactive oxygen species accelerate susceptibility to experimental thrombosis in vivo. APPROACH AND RESULTS: Using a murine model with genetic deficiency in superoxide dismutase-1 (SOD1), we measured susceptibility to carotid artery thrombosis in response to photochemical injury. We found that SOD1-deficient (Sod1(-/-)) mice formed stable arterial occlusions significantly faster than wild-type (Sod1(+/+)) mice (P<0.05). Sod1(-/-) mice also developed significantly larger venous thrombi than Sod1(+/+) mice after inferior vena cava ligation (P<0.05). Activation of protein C by thrombin in lung was diminished in Sod1(-/-) mice (P<0.05 versus Sod1(+/+) mice), and generation of activated protein C in response to infusion of thrombin in vivo was decreased in Sod1(-/-) mice (P<0.05 versus Sod1(+/+) mice). SOD1 deficiency had no effect on the expression of thrombomodulin, endothelial protein C receptor, or tissue factor in lung or levels of protein C in plasma. Exposure of human thrombomodulin to superoxide in vitro caused oxidation of multiple methionine residues, including critical methionine 388, and a 40% decrease in thrombomodulin-dependent activation of protein C (P<0.05). SOD and catalase protected against superoxide-induced methionine oxidation and restored protein C activation in vitro (P<0.05). CONCLUSIONS: SOD prevents thrombomodulin methionine oxidation, promotes protein C activation, and protects against arterial and venous thrombosis in mice.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Proteína C/metabolismo , Superóxido Dismutasa/deficiencia , Trombosis/enzimología , Trombosis de la Vena/enzimología , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Catalasa/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Ratones de la Cepa 129 , Ratones Transgénicos , Oxidación-Reducción , Estrés Oxidativo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Superóxidos/metabolismo , Trombomodulina/metabolismo , Trombosis/genética , Trombosis/patología , Vena Cava Inferior/enzimología , Vena Cava Inferior/patología , Trombosis de la Vena/patología
19.
Atherosclerosis ; 241(1): 111-20, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25974101

RESUMEN

In vitro, insulin has mitogenic effects on vascular smooth muscle cells (VSMC) but also has protective effects on endothelial cells by stimulating nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) expression. Furthermore, NOS inhibition attenuates the effect of insulin to inhibit VSMC migration in vitro. Using an in vivo model, we have previously shown that insulin decreases neointimal growth and cell migration and increases re-endothelialization after arterial injury in normal rats. Since insulin can stimulate NOS, and NO can decrease neointimal growth, we hypothesized that NOS, and more specifically eNOS was required for the effects of insulin in vivo. Rats were given subcutaneous insulin implants (3 U/day) alone or with the NOS inhibitor l-NAME (2 mg kg(-1) day(-1)) 3 days before arterial (carotid or aortic) balloon catheter injury. Insulin decreased both neointimal area (P < 0.01) and cell migration (P < 0.01), and increased re-endothelialization (P < 0.05). All of these effects were prevented by the co-administration of l-NAME. Insulin was found to decrease inducible NOS expression (P < 0.05) but increase eNOS phosphorylation (P < 0.05). These changes were also translated at the functional level where insulin improved endothelial-dependent vasorelaxation. To further study the NOS isoform involved in insulin action, s.c. insulin (0.1 U/day) was given to wild-type and eNOS knockout mice. We found that insulin was effective at decreasing neointimal formation in wild-type mice after wire injury of the femoral artery, whereas this effect of insulin was absent in eNOS knockout mice. These results show that the vasculoprotective effect of insulin after arterial injury is mediated by an eNOS-dependent mechanism.


Asunto(s)
Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Insulina/administración & dosificación , Neointima , Óxido Nítrico Sintasa de Tipo III/metabolismo , Lesiones del Sistema Vascular/tratamiento farmacológico , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/enzimología , Aorta Torácica/lesiones , Aorta Torácica/patología , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Inhibidores Enzimáticos/farmacología , Arteria Femoral/efectos de los fármacos , Arteria Femoral/enzimología , Arteria Femoral/lesiones , Arteria Femoral/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Ratas Sprague-Dawley , Repitelización/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Lesiones del Sistema Vascular/enzimología , Lesiones del Sistema Vascular/patología , Lesiones del Sistema Vascular/fisiopatología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
20.
Circulation ; 131(13): 1191-201, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25712208

RESUMEN

BACKGROUND: ADAMTS-7, a member of the disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family, was recently identified to be significantly associated genomewide with coronary artery disease. However, the mechanisms that link ADAMTS-7 and coronary artery disease risk remain elusive. We have previously demonstrated that ADAMTS-7 promotes vascular smooth muscle cell migration and postinjury neointima formation via degradation of a matrix protein cartilage oligomeric matrix protein. Because delayed endothelium repair renders neointima and atherosclerosis plaque formation after vessel injury, we examined whether ADAMTS-7 also inhibits re-endothelialization. METHODS AND RESULTS: Wire injury of the carotid artery and Evans blue staining were performed in Adamts7(-/-) and wild-type mice. Adamts-7 deficiency greatly promoted re-endothelialization at 3, 5, and 7 days after injury. Consequently, Adamts-7 deficiency substantially ameliorated neointima formation in mice at days 14 and 28 after injury in comparison with the wild type. In vitro studies further indicated that ADAMTS-7 inhibited both endothelial cell proliferation and migration. Surprisingly, cartilage oligomeric matrix protein deficiency did not affect endothelial cell proliferation/migration and re-endothelialization in mice. In a further examination of other potential vascular substrates of ADAMTS-7, a label-free liquid chromatography-tandem mass spectrometry secretome analysis revealed thrombospondin-1 as a potential ADAMTS-7 target. The subsequent studies showed that ADAMTS-7 was directly associated with thrombospondin-1 by its C terminus and degraded thrombospondin-1 in vivo and in vitro. The inhibitory effect of ADAMTS-7 on postinjury endothelium recovery was circumvented in Tsp1(-/-) mice. CONCLUSIONS: Our study revealed a novel mechanism by which ADAMTS-7 affects neointima formation. Thus, ADAMTS-7 is a promising treatment target for postinjury vascular intima hyperplasia.


Asunto(s)
Proteínas ADAM/fisiología , Traumatismos de las Arterias Carótidas/enzimología , Arteria Femoral/lesiones , Neointima/enzimología , Trombospondina 1/fisiología , Remodelación Vascular/fisiología , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Proteína ADAMTS7 , Secuencia de Aminoácidos , Animales , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , División Celular , Células Cultivadas , Células Endoteliales/metabolismo , Arteria Femoral/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Miocitos del Músculo Liso/metabolismo , Neointima/patología , Mapeo de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Trombospondina 1/deficiencia , Trombospondina 1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...