Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 10(30): e2303283, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37667861

RESUMEN

Myocardial infarction (MI) remains the leading cause of death worldwide. Cardiac fibroblasts (CFs) are abundant in the heart and are responsible for cardiac repair post-MI. NF-κB-repressing factor (NKRF) plays a significant role in the transcriptional inhibition of various specific genes. However, the NKRF action mechanism in CFs remains unclear in cardiac repair post-MI. This study investigates the NKRF mechanism in cardiac remodeling and dysfunction post-MI by establishing a CF-specific NKRF-knockout (NKRF-CKO) mouse model. NKRF expression is downregulated in CFs in response to pathological cardiac remodeling in vivo and TNF-α in vitro. NKRF-CKO mice demonstrate worse cardiac function and survival and increased infarct size, heart weight, and MMP2 and MMP9 expression post-MI compared with littermates. NKRF inhibits CF migration and invasion in vitro by downregulating MMP2 and MMP9 expression. Mechanistically, NKRF inhibits human antigen R (HuR) transcription by binding to the classical negative regulatory element within the HuR promoter via an NF-κB-dependent mechanism. This decreases HuR-targeted Mmp2 and Mmp9 mRNA stability. This study suggests that NKRF is a therapeutic target for pathological cardiac remodeling.


Asunto(s)
Infarto del Miocardio , FN-kappa B , Animales , Humanos , Ratones , Fibroblastos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Infarto del Miocardio/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/genética , Remodelación Ventricular/genética
2.
Circ Res ; 129(12): 1141-1157, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34704457

RESUMEN

RATIONALE: The NLRP3 (NLR [NOD-like receptor] family, pyrin domain containing 3) inflammasome is an important driver of atherosclerosis. Our previous study shows that chaperone-mediated autophagy (CMA), one of the main lysosomal degradative process, has a regulatory role in lipid metabolism of macrophages. However, whether the NLRP3 inflammasome is regulated by CMA, and the role of CMA in atherosclerosis remains unclear. OBJECTIVE: To determine the role of CMA in the regulation of NLRP3 inflammasome and atherosclerosis. METHODS AND RESULTS: The expression of CMA marker, LAMP-2A (lysosome-associated membrane protein type 2A), was first analyzed in ApoE-/- mouse aortas and human coronary atherosclerotic plaques, and a significant downregulation of LAMP-2A in advanced atherosclerosis in both mice and humans was observed. To selectively block CMA, we generated macrophage-specific conditional LAMP-2A knockout mouse strains in C57BL/6 mice and ApoE-/- mice. Deletion of macrophage LAMP-2A accelerated atherosclerotic lesion formation in the aortic root and the whole aorta in ApoE-/- mice. Mechanistically, LAMP-2A deficiency promoted NLRP3 inflammasome activation and subsequent release of mature IL (interleukin)-1ß in macrophages and atherosclerotic plaques. Furthermore, gain-of-function studies verified that restoration of LAMP-2A levels in LAMP-2A-deficient macrophages greatly attenuated NLRP3 inflammasome activation. Importantly, we identified the NLRP3 protein as a CMA substrate and demonstrated that LAMP-2A deficiency did not affect the NLRP3 mRNA levels but hindered degradation of the NLRP3 protein through CMA pathway. CONCLUSIONS: CMA function becomes impaired during the progression of atherosclerosis, which increases NLRP3 inflammasome activation and secretion of IL-1ß, promoting vascular inflammation and atherosclerosis progression. Our study unveils a new mechanism by which NLRP3 inflammasome is regulated in macrophages and atherosclerosis, thus providing a new insight into the role of autophagy-lysosomal pathway in atherosclerosis. Pharmacological activation of CMA may provide a novel therapeutic strategy for atherosclerosis and other NLRP3 inflammasome/IL-1ß-driven diseases.


Asunto(s)
Aterosclerosis/metabolismo , Autofagia , Inflamasomas/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Interleucina-1beta/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética
3.
J Cardiovasc Pharmacol ; 73(4): 223-231, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30540688

RESUMEN

Atorvastatin is a lipid-regulating drug that is commonly used in clinical practice and can stabilize plaques. Increasing evidence shows that statins have anti-heart failure (HF) effects, but their specific mechanism is not clear. The purpose of this study was to investigate the cardioprotective effects of atorvastatin on HF in rats and its mechanism. Continuous intraperitoneal injection of 2.5 mg/kg/w doxorubicin for 6 weeks, with a cumulative dose of 15 mg/kg, was used to induce a rat model of HF. Then, the rats were treated with low-dose atorvastatin, high-dose atorvastatin, or saline for 4 weeks. In the DOX-treated groups, echocardiography showed decreases in left ventricular ejection fraction and fractional shortening and increases in left ventricular end-diastolic diameter and left ventricular posterior wall thickness compared with those in the control group, and increased levels of brain natriuretic peptide and Hsp70 were also found in the doxorubicin-treated groups. Compared with saline intervention, atorvastatin ameliorated left ventricular ejection fraction, fractional shortening, left ventricular end-diastolic diameter, and left ventricular posterior wall thickness (a significant difference was observed only in the high-dose group) and reduced serum brain natriuretic peptide. Hematoxylin and eosin staining showed that atorvastatin ameliorated myocardial injury. The improvement in cardiac function induced by atorvastatin was accompanied by increased Hsp70 expression, decreased p-ERK and p-JNK expression, and a reduction in myocardial fibrosis shown by Masson staining. In addition, atorvastatin had a protective effect on the myocardial apoptosis signaling pathway, with increased p-Akt expression and downregulated cleaved caspase-3 expression, and the reduction in myocardial apoptosis was confirmed by a TUNEL assay. Therefore, our experiments demonstrated that atorvastatin may protect cardiac function by modulating Hsp70, p-Akt, p-ERK, and p-JNK signaling to reduce myocardial fibrosis and myocardial apoptosis.


Asunto(s)
Atorvastatina/farmacología , Doxorrubicina , Proteínas HSP70 de Choque Térmico/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Ventrículos Cardíacos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Disfunción Ventricular Izquierda/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Cardiotoxicidad , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Masculino , Fosforilación , Ratas Wistar , Transducción de Señal , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/efectos de los fármacos
4.
Biosci Rep ; 38(5)2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30185439

RESUMEN

There is increasing evidence that serum adipokine levels are associated with higher risks of cardiovascular diseases. As an important adipokine, fibroblast growth factor 21 (FGF21) has been demonstrated to be associated with atherosclerosis and coronary artery disease (CAD). However, circulating level of FGF21 in patients with angina pectoris has not yet been investigated. Circulating FGF21 level was examined in 197 patients with stable angina pectoris (SAP, n=66), unstable angina pectoris (UAP, n=76), and control subjects (n=55) along with clinical variables of cardiovascular risk factors. Serum FGF21 concentrations on admission were significantly increased more in patients with UAP than those with SAP (Ln-FGF21: 5.26 ± 0.87 compared with 4.85 ± 0.77, P<0.05) and control subjects (natural logarithm (Ln)-FGF21: 5.26 ± 0.87 compared with 4.54 ± 0.72, P<0.01). The correlation analysis revealed that serum FGF21 concentration was positively correlated with the levels of cardiac troponin I (cTnI) (r2 = 0.026, P=0.027) and creatine kinase-MB (CK-MB) (r2 = 0.023, P= 0.04). Furthermore, FGF21 level was identified as an independent factor associated with the risks of UAP (odds ratio (OR): 2.781; 95% CI: 1.476-5.239; P=0.002), after adjusting for gender, age, and body mass index (BMI). However, there were no correlations between serum FGF21 levels and the presence of SAP (OR: 1.248; 95% CI: 0.703-2.215; P=0.448). The present study indicates that FGF21 has a strong correlation and precise predictability for increased risks of UAP, that is independent of traditional risk factors of angina pectoris.


Asunto(s)
Angina Estable/genética , Angina Inestable/genética , Enfermedad de la Arteria Coronaria/genética , Factores de Crecimiento de Fibroblastos/genética , Anciano , Angina Estable/sangre , Angina Estable/fisiopatología , Angina Inestable/sangre , Angina Inestable/fisiopatología , Aterosclerosis/sangre , Aterosclerosis/genética , Índice de Masa Corporal , Enfermedad de la Arteria Coronaria/fisiopatología , Femenino , Factores de Crecimiento de Fibroblastos/sangre , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , Troponina I/sangre
5.
PLoS One ; 13(8): e0201395, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30125282

RESUMEN

Various angiogenic factors have been shown to play important roles in intraplaque angiogenesis, while little is known about the dynamic expression change and interplay between various angiogenic factors and intraplaque angiogenesis under high cholesterol conditions. New Zealand rabbits underwent balloon injury of the abdominal artery and then were assigned to a control group (n = 15, normal chow) or high cholesterol group (n = 25, 1% high cholesterol diet). At weeks 4, 6, 8, 10, and 12 after acclimation, rabbits (high cholesterol group, n = 5; control group, n = 3) were euthanized. No lesions were observed in the control group. From week 4 to week 12, the expression of vascular endothelial growth factor A (VEGF-A), VEGF receptor 2 (VEGFR-2), fibroblast growth factor 2 (FGF-2), FGF receptor 1 (FGFR-1), platelet-derived growth factor-BB (PDGF-BB), and tumor necrosis factor alpha (TNF-α), the vulnerability index (VI) and the microvessel density (MVD) were significantly elevated in the high cholesterol group; however, PDGF receptor ß (PDGFR-ß) expression showed little change. Analysis by double-label immunofluorescence (CD31 and Ng2) and FITC-dextran indicated that the neovessels within the plaque were leaky due to a lack of pericytes. As indicated by Pearson's correlation analysis, there was a highly positive correlation between the VI, MVD, macrophage content, and TNF-α level, and the levels of VEGF-A/VEGFR-2 and FGF-2/FGFR-1. However, no correlations were observed between PDGFR-ß levels and the VI or MVD. High expression of VEGF-A/VEGFR-2 and FGF-2/FGFR-1 but not of PDGF-BB/PDGFR-ß may contribute to immature and inflammatory intraplaque angiogenesis and plaque instability in a rabbit model of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Neovascularización Patológica/metabolismo , Placa Aterosclerótica/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Animales , Aterosclerosis/patología , Becaplermina/biosíntesis , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/patología , Masculino , Neovascularización Patológica/patología , Pericitos/metabolismo , Pericitos/patología , Placa Aterosclerótica/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Conejos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 38(2): 425-437, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29284609

RESUMEN

OBJECTIVE: DKK3 (dickkopf 3), a 36-kD secreted glycoprotein, has been shown to be involved in the differentiation of partially reprogrammed cells and embryonic stem cells to smooth muscle cells (SMCs), but little is known about its involvement in vascular disease. This study aims to assess the effects of DKK3 on atherosclerotic plaque composition. APPROACH AND RESULTS: In the present study, we used a murine model of atherosclerosis (ApoE-/-) in conjunction with DKK3-/- and performed tandem stenosis of the carotid artery to evaluate atherosclerotic plaque development. We found that the absence of DKK3 leads to vulnerable atherosclerotic plaques, because of a reduced number of SMCs and reduced matrix protein deposition, as well as increased hemorrhage and macrophage infiltration. Further in vitro studies revealed that DKK3 can induce differentiation of Sca1+ (stem cells antigen 1) vascular progenitors and fibroblasts into SMCs via activation of the TGF-ß (transforming growth factor-ß)/ATF6 (activating transcription factor 6) and Wnt signaling pathways. Finally, we assessed the therapeutic potential of DKK3 in mouse and rabbit models and found that DKK3 altered the atherosclerotic plaque content via increasing SMC numbers and reducing vascular inflammation. CONCLUSIONS: Cumulatively, we provide the first evidence that DKK3 is a potent SMC differentiation factor, which might have a therapeutic effect in reducing intraplaque hemorrhage related to atherosclerotic plaque phenotype.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Estenosis Carotídea/metabolismo , Transdiferenciación Celular , Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica , Células Madre/metabolismo , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Ataxina-1/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Estenosis Carotídea/genética , Estenosis Carotídea/patología , Células Cultivadas , Quimiocinas , Modelos Animales de Enfermedad , Femenino , Fibroblastos/patología , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/patología , Hemorragia/prevención & control , Humanos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fenotipo , Conejos , Células Madre/patología , Factor de Crecimiento Transformador beta1/metabolismo , Vía de Señalización Wnt
7.
J Mol Cell Cardiol ; 114: 243-252, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29196099

RESUMEN

AIMS: Cardiac pressure and humoral factors induce cardiac hypertrophy and fibrosis, which are characterized by increased stiffness, reduced contractility and altered perfusion. Angiotensin II (AngII) is well known to promote this pathology. Angiotensin-converting enzyme (ACE) 2, which cleaves AngII and forms Ang-(1-7), exerts protective anti-hypertrophy and anti-fibrosis effects. A disintegrin and metalloproteinase 17 (ADAM17), a membrane-bound enzyme reported to cleave ACE2, may participate in the pathological process of AngII perfusion-induced heart damage. However, researchers have not clearly determined whether dickkopf-3 (DKK3) regulates the ADAM17/ACE2 pathway and, if so, whether DKK3-mediated regulation is related to the glycogen synthase kinase-3ß (GSK-3ß)/ß-catenin pathway. In this study, we explored whether DKK3 overexpression ameliorates the development of AngII-induced cardiac fibrosis and hypertrophy through the ADAM17/ACE2 and GSK-3ß/ß-catenin pathways. METHODS: Mice were injected with a DKK3-overexpressing adenovirus or vehicle and then infused with AngII or saline using subcutaneously implanted mini-pumps for four weeks. Hearts were stained with hematoxylin-eosin, Masson's trichrome and immunohistochemical markers for histology. Primary fibroblasts were treated with the adenovirus and AngII and then examined using western blotting, EdU (5-ethynyl-2'-deoxyuridine) assays and immunofluorescence. Additionally, siRNA silencing was performed to study the role of DKK3 and the involved pathways. RESULTS: AngII-induced cardiac hypertrophy and interstitial and perivascular fibrosis were less severe in DKK3-overexpressing mice than in control mice. Moreover, the expression levels of fibrotic genes, such as collagen I and III, and the hypertrophic genes atrial natriuretic peptide (ANP) and beta-myosin heavy chain (ß-MHC) were decreased. DKK3 overexpression also exerted a protective effect by inhibiting ADAM17 phosphorylation, thus increasing ACE2 expression and subsequently promoting AngII degradation. Furthermore, this process was mediated by the inhibition of GSK-3ß and ß-catenin and decreased translocation of ß-catenin to the nucleus. On the other hand, the DKK3 knockdown by siRNA achieved opposite results. CONCLUSION: DKK3 overexpression substantially alleviated AngII infusion-induced cardiac hypertrophy and fibrosis by regulating ADAM17/ACE2 pathway activity and inhibiting the GSK-3ß/ß-catenin pathway.


Asunto(s)
Proteína ADAM17/metabolismo , Angiotensina II/farmacología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Angiotensina I , Enzima Convertidora de Angiotensina 2 , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Cardiomegalia/fisiopatología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Inflamación/patología , Metaloproteinasas de la Matriz/metabolismo , Ratones Endogámicos C57BL , Fragmentos de Péptidos , Peptidil-Dipeptidasa A/metabolismo , Perfusión , Fosforilación/efectos de los fármacos , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
8.
J Cell Mol Med ; 22(3): 1475-1488, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29266779

RESUMEN

Diabetic cardiomyopathy, a major cardiac complication, contributes to heart remodelling and heart failure. Our previous study discovered that CCAAT/enhancer-binding protein ß (C/EBPß), a transcription factor that belongs to a family of basic leucine zipper transcription factors, interacts with the angiotensin-converting enzyme 2 (ACE2) promoter sequence in other disease models. Here, we aimed to determine the role of C/EBPß in diabetes and whether ACE2 expression is regulated by C/EBPß. A type 1 diabetic mouse model was generated by an intraperitoneal injection of streptozotocin. Diabetic mice were injected with a lentivirus expressing either C/EBPß or sh-C/EBPß or treated with valsartan after 12 weeks to observe the effects of C/EBPß. In vitro, cardiac fibroblasts and cardiomyocytes were treated with high glucose (HG) to investigate the anti-fibrosis, anti-apoptosis and regulatory mechanisms of C/EBPß. C/EBPß expression was down-regulated in diabetic mice and HG-induced cardiac neonatal cells. C/EBPß overexpression significantly attenuated collagen deposition and cardiomyocyte apoptosis by up-regulating ACE2 expression. The molecular mechanism involved the binding of C/EBPß to the ACE2 promoter sequence. Although valsartan, a classic angiotensin receptor blocker, relieved diabetic complications, the up-regulation of ACE2 expression by C/EBPß overexpression may exert greater beneficial effects on patients with diabetic cardiomyopathy.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Diabetes Mellitus Experimental/terapia , Cardiomiopatías Diabéticas/prevención & control , Fibroblastos/metabolismo , Miocitos Cardíacos/metabolismo , Peptidil-Dipeptidasa A/genética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Apoptosis/genética , Glucemia/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular , Colágeno/antagonistas & inhibidores , Colágeno/genética , Colágeno/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica , Glucosa/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Peptidil-Dipeptidasa A/metabolismo , Cultivo Primario de Células , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Estreptozocina , Valsartán/farmacología
9.
Front Pharmacol ; 8: 727, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29114222

RESUMEN

Atherosclerosis (AS) is a lipid-driven disease in which macrophage foam cells play a critical role by increasing vascular lipid accumulation and contributing to plaque instability. Ginsenoside Rb1 (Rb1), the most abundant active component of ginseng, has been found potently to promote lipid metabolism and attenuate lipid accumulation. However, the underlying mechanisms remain unclear. In this study, the effects of Rb1 on lipid accumulation and plaque stability were investigated both in vitro and in vivo by using primary peritoneal macrophages isolated from C57BL/6 mice and an AS model in ApoE-/- mice. The results showed that Rb1 reduced lipid accumulation both in macrophage foam cells and atherosclerotic plaques. Rb1 treatment promoted plaque stability by modifying plaque composition via the activation of autophagy both in vitro and in vivo. Transmission electron microscopy further showed an increased accumulation of autophagolysosomes in Rb1-treated macrophage foam cells. However, the modulation of lipid accumulation by Rb1 was attenuated by autophagy blockage using autophagy-related gene 5 (Atg5) small interfering RNA (siRNA) in vitro. In addition, Rb1 notably increased AMPK phosphorylation both in vitro and in vivo, and the AMPK inhibitor compound C abolished the Rb1-induced autophagy in macrophage foam cells. In conclusion, ginsenoside Rb1 reduced lipid accumulation in macrophage foam cells and enhanced atherosclerotic plaque stability by the induction of macrophage autophagy. Our study provides new evidence for the possible use of Rb1 in the prevention and treatment of AS.

11.
Coron Artery Dis ; 26(4): 322-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25768243

RESUMEN

BACKGROUND: Coronary atherosclerotic plaque formation is driven by macrophage infiltration. Monocytes and macrophages contribute to the progression of atherosclerosis. However, research on the relationship between AMP-activated protein kinase (AMPK) and vulnerable atherosclerotic plaques is still insufficient. In the present study, we aimed to elucidate the adaptive mechanism between autophagy of peripheral blood monocytes (PBMs) and the rupture of atherosclerotic plaques. We investigated whether AMPK and autophagy of monocytes can enhance the stability of coronary atherosclerotic plaques in the human body. PATIENTS AND METHODS: Samples of PBMs were collected and isolated from all patients with stable angina pectoris (SAP), non-ST-segment elevation acute coronary syndrome, ST-segment elevation acute myocardial infarction, and without coronary artery disease (control). Then, western blot was used to detect the expression levels of AMPK and autophagy-related protein. RESULTS: The expression levels of beclin-1 and ATG7 were all significantly lower in the acute coronary syndrome groups than those in the SAP and control groups (all P<0.01). The level of phosphorylated AMPK was significantly decreased in patients with acute coronary syndrome compared with those in the SAP and control groups (P<0.01). However, there was no statistical difference between the SAP group and the control group. The activation of mTOR was distinctly increased in the STEMI group (P<0.05). CONCLUSION: Therefore, our work is novel in showing that AMPK of PBMs may decrease plaque vulnerability and subsequent plaque rupture through activation of autophagy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Síndrome Coronario Agudo/patología , Angina de Pecho/patología , Autofagia , Enfermedad de la Arteria Coronaria/patología , Monocitos/patología , Infarto del Miocardio/patología , Síndrome Coronario Agudo/sangre , Adulto , Anciano , Anciano de 80 o más Años , Angina de Pecho/sangre , Proteínas Reguladoras de la Apoptosis/sangre , Proteína 7 Relacionada con la Autofagia , Beclina-1 , Western Blotting , Enfermedad de la Arteria Coronaria/enzimología , Electrocardiografía , Femenino , Humanos , Masculino , Proteínas de la Membrana/sangre , Persona de Mediana Edad , Infarto del Miocardio/sangre , Enzimas Activadoras de Ubiquitina/sangre
12.
PLoS One ; 9(3): e90563, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24599185

RESUMEN

Macrophage infiltration contributes to the instability of atherosclerotic plaques. In the present study, we investigated whether selective inhibition of PI3K/Akt/mTOR signaling pathway can enhance the stability of atherosclerotic plaques by activation of macrophage autophagy. In vitro study, selective inhibitors or siRNA of PI3K/Akt/mTOR pathways were used to treat the rabbit's peritoneal primary macrophage cells. Inflammation related cytokines secreted by macrophages were measured. Ultrastructure changes of macrophages were examined by transmission electron microscope. mRNA or protein expression levels of autophagy related gene Beclin 1, protein 1 light chain 3 II dots (LC3-II) or Atg5-Atg12 conjugation were assayed by quantitative RT-PCR or Western blot. In vivo study, vulnerable plaque models were established in 40 New Zealand White rabbits and then drugs or siRNA were given for 8 weeks to inhibit the PI3K/Akt/mTOR signaling pathway. Intravascular ultrasound (IVUS) was performed to observe the plaque imaging. The ultrastructure of the abdominal aortic atherosclerosis lesions were analyzed with histopathology. RT-PCR or Western blot methods were used to measure the expression levels of corresponding autophagy related molecules. We found that macrophage autophagy was induced in the presence of Akt inhibitor, mTOR inhibitor and mTOR-siRNA in vitro study, while PI3K inhibitor had the opposite role. In vivo study, we found that macrophage autophagy increased significantly and the rabbits had lower plaque rupture incidence, lower plaque burden and decreased vulnerability index in the inhibitors or siRNA treated groups. We made a conclusion that selective inhibition of the Akt/mTOR signal pathway can reduce macrophages and stabilize the vulnerable atherosclerotic plaques by promoting macrophage autophagy.


Asunto(s)
Macrófagos Peritoneales/fisiología , Placa Aterosclerótica/tratamiento farmacológico , Transducción de Señal , Animales , Aterosclerosis/sangre , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Autofagia , Células Cultivadas , Cromonas/farmacología , Citocinas/metabolismo , Lípidos/sangre , Macrófagos Peritoneales/efectos de los fármacos , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Placa Aterosclerótica/sangre , Placa Aterosclerótica/inmunología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Conejos , Ribonucleósidos/farmacología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA