Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 323
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 1315: 205-236, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34302694

RESUMEN

Hydrogen sulfide (H2S), nitric oxide (NO), carbon monoxide (CO), and sulfur dioxide (SO2) were previously considered as toxic gases, but now they are found to be members of mammalian gasotransmitters family. Both H2S and SO2 are endogenously produced in sulfur-containing amino acid metabolic pathway in vivo. The enzymes catalyzing the formation of H2S are mainly CBS, CSE, and 3-MST, and the key enzymes for SO2 production are AAT1 and AAT2. Endogenous NO is produced from L-arginine under catalysis of three isoforms of NOS (eNOS, iNOS, and nNOS). HO-mediated heme catabolism is the main source of endogenous CO. These four gasotransmitters play important physiological and pathophysiological roles in mammalian cardiovascular, nervous, gastrointestinal, respiratory, and immune systems. The similarity among these four gasotransmitters can be seen from the same and/or shared signals. With many studies on the biological effects of gasotransmitters on multiple systems, the interaction among H2S and other gasotransmitters has been gradually explored. H2S not only interacts with NO to form nitroxyl (HNO), but also regulates the HO/CO and AAT/SO2 pathways. Here, we review the biosynthesis and metabolism of the gasotransmitters in mammals, as well as the known complicated interactions among H2S and other gasotransmitters (NO, CO, and SO2) and their effects on various aspects of cardiovascular physiology and pathophysiology, such as vascular tension, angiogenesis, heart contractility, and cardiac protection.


Asunto(s)
Gasotransmisores , Sulfuro de Hidrógeno , Animales , Monóxido de Carbono , Mamíferos , Óxido Nítrico , Dióxido de Azufre
2.
Pharmacol Res ; 159: 104926, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32502636

RESUMEN

Cardiac remodeling is accompanied by cardiac hypertrophy, fibrosis, dysfunction, and eventually leading to heart failure. Intermedin (IMD), as a paracrine/autocrine peptide, has a protective effect in cardiovascular diseases. In this study, we elucidated the role and the underlying mechanism of IMD in pathological remodeling. Pathological remodeling mouse models were induced by abdominal aorta constriction for 4 weeks or angiotensin II (Ang II) infusion for 2 weeks in wildtype, IMD-overexpression, IMD-knockout and klotho-knockdown mice. Western blot, real-time PCR, histological staining, echocardiography and hemodynamics were used to detect the role of IMD in cardiac remodeling. Cardiac hypertrophy, fibrosis and dysfunction were significantly aggravated in IMD-knockout mice versus wildtype mice, and the expression of klotho was downregulated. Conversely, cardiac remodeling was alleviated in IMD-overexpression mice, and the expression of klotho was upregulated. Hypertension induced by Ang II infusion rather than abdominal aorta constriction was mitigated by IMD. However, the cardioprotective effect of IMD was blocked in klotho-knockdown mice. Similar results were found in cultured neonatal rat cardiomyocytes, which was pretreated with IMD before Ang II stimulation. Mechanistically, IMD inhibited the phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and the activity of calcineurin to protect against cardiac hypertrophy through upregulating klotho in vivo and in vitro. Furthermore, peroxisome proliferator-activated receptor γ (PPARγ) might mediate IMD upregulating klotho. In conclusion, pathological remodeling may be alleviated by endogenous IMD, which inhibits the expression of calcineurin and p-CaMKII by upregulating klotho via the PPARγ pathway. It suggested that IMD might be a therapeutic target for heart disease.


Asunto(s)
Glucuronidasa/metabolismo , Hipertrofia Ventricular Izquierda/prevención & control , Miocitos Cardíacos/metabolismo , Neuropéptidos/metabolismo , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda , Remodelación Ventricular , Angiotensina II , Animales , Aorta Abdominal/fisiopatología , Aorta Abdominal/cirugía , Calcineurina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Constricción , Modelos Animales de Enfermedad , Fibrosis , Glucuronidasa/genética , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Proteínas Klotho , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Neuropéptidos/genética , PPAR gamma/metabolismo , Hormonas Peptídicas/farmacología , Fosforilación , Ratas Sprague-Dawley , Transducción de Señal , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
3.
J Cell Physiol ; 234(10): 17578-17588, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30793300

RESUMEN

Cardiac hypertrophy is the main cause of heart failure and sudden death in patients. But the pathogenesis is unclear. Angiotensin II may contribute to cardiac hypertrophy in response to pressure overload. In angiotensin II-treated cardiomyocytes, there is a larger cross-sectional area, more apoptosis cells, and a reduction of irisin expression. An increase in P62, an autophagy flux index, as well as LC3II, were observed in cardiomyocytes after angiotensin II-induced injury. Surprisely, irisin supplementation increased LC3II expression and decreased P62 expression, consisted of results of RFP-GFP-LC3B adenovirus transfection, and reduced cardiomyocyte apoptosis, meanwhile, the protection of irisin was reversed by the autophagy inhibitor 3-methyladenine. In animal experiments, overexpression of irisin reduced cardiomyocyte apoptosis and alleviated myocardial hypertrophy caused by pressure overload. The above results indicate that irisin-induced protective autophagy and alleviated the apoptosis signaling pathway in cardiomyocytes, consequently reducing cardiomyocyte apoptosis after angiotensin II-induced injury. Hence, increasing irisin expression may be a new way to improve cardiac function and quality of life in patients with cardiac hypertrophy.


Asunto(s)
Angiotensina II/fisiología , Apoptosis/fisiología , Autofagia/fisiología , Fibronectinas/farmacología , Fibronectinas/fisiología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Angiotensina II/farmacología , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Cardiomegalia/etiología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Células Cultivadas , Regulación hacia Abajo , Fibronectinas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
4.
Int Heart J ; 60(1): 45-49, 2019 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-30393265

RESUMEN

Neuregulin-4 (Nrg4) is a newly discovered adipokine that is synthesized in many tissues and plays an important role in modulating systemic energy metabolism and in the development of metabolic disorders. However, little is known about the relationship between Nrg4 and coronary artery disease (CAD). In this study, we investigated the association between Nrg4 and the presence and severity of CAD.We enrolled 73 patients diagnosed by coronary angiography (CAG) as having CAD and 32 controls. The CAD group was divided into two subgroups according to their SYNTAX score. Plasma levels of Nrg4 were measured in all participants and compared among different groups. The relationship between Nrg4 and CAD was analyzed. Receiver operating characteristic (ROC) analysis was conducted to evaluate the usefulness Nrg4 in assessing the presence and severity of CAD.Nrg4 levels were negatively associated with the SYNTAX score (r = -0.401, P = 0.000). The patients with a higher SYNTAX score had significantly lower Nrg4 levels as compared with the low SYNTAX score subgroup and the controls (P < 0.05). The Nrg4 levels of the low SYNTAX score subgroup were much lower than controls (P < 0.05). Furthermore, an association between Nrg4 and CAD (odds ratio, 0.279; 95% confidence interval, 0.088-0.882) was observed. Nrg4 had 43.8% sensitivity and 96.9% specificity for identifying CAD, and 73.1% sensitivity and 87.3% specificity for identifying patients who had severe coronary artery lesions.Nrg4 levels were found to be inversely associated with the presence and severity of CAD.


Asunto(s)
Biomarcadores/sangre , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Neurregulinas/sangre , Anciano , Estudios de Casos y Controles , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Curva ROC , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad
5.
J Mol Cell Cardiol ; 121: 242-255, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30053525

RESUMEN

In hypertrophic hearts, autophagic flux insufficiency is recognized as a key pathology leading to maladaptive cardiac remodeling and heart failure. This study aimed to illuminate the cardioprotective role and mechanisms of a new myokine and adipokine, irisin, in cardiac hypertrophy and remodeling. Adult male wild-type, mouse-FNDC5 (irisin-precursor)-knockout and FNDC5 transgenic mice received 4 weeks of transverse aortic constriction (TAC) alone or combined with intraperitoneal injection of chloroquine diphosphate (CQ). Endogenous FNDC5 ablation aggravated and exogenous FNDC5 overexpression attenuated the TAC-induced hypertrophic damage in the heart, which was comparable to the protection of irisin against cardiomyocyte hypertrophy induced by angiotensin II (Ang II) or phenylephrine (PE). Accumulated autophagosome and impaired autophagy flux occurred in the TAC-treated myocardium and Ang II- or PE-insulted cardiomyocytes. Irisin deficiency caused reduced autophagy and aggravated autophagy flux failure, whereas irisin overexpression or supplementation induced protective autophagy and improved autophagy flux, which were reversed by autophagy inhibitors Atg5 siRNA, 3-MA and CQ. Irisin boosted the activity of only AMPK but not Akt and MAPK family members in hypertrophic hearts and cultured cardiomyocytes and further activated ULK1 at Ser555 but not Ser757 and did not affect the mTOR-S6K axis. Blockage of AMPK and ULK1 with compund C and SBI-0206965, respectively, both abrogated irisin's protection against cardiomyocyte hypertrophic injury and reversed its induction of both autophagy and autophagy flux. Our results suggest that irisin protects against pressure overload-induced cardiac hypertrophy by inducing protective autophagy and autophagy flux via activating AMPK-ULK1 signaling.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Cardiomegalia/genética , Fibronectinas/genética , Insuficiencia Cardíaca/genética , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Angiotensina II/administración & dosificación , Animales , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Benzamidas/administración & dosificación , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/patología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/efectos de los fármacos , Fenilefrina/administración & dosificación , Presión , Pirimidinas/administración & dosificación , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
6.
Arterioscler Thromb Vasc Biol ; 36(11): 2176-2190, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27634835

RESUMEN

OBJECTIVE: Oxidative stress plays a critical role in the development of abdominal aortic aneurysm (AAA). Intermedin (IMD) is a regulator of oxidative stress. Here, we investigated whether IMD reduces AAA by inhibiting oxidative stress. APPROACH AND RESULTS: In angiotensin II-induced ApoE-/- mouse and CaCl2-induced C57BL/6J mouse model of AAA, IMD1-53 significantly reduced the incidence of AAA and maximal aortic diameter. Ultrasonography, hematoxylin, and eosin staining and Verhoeff-van Gieson staining showed that IMD1-53 significantly decreased the enlarged aortas and elastic lamina degradation induced by angiotensin II or CaCl2. Mechanistically, IMD1-53 attenuated oxidative stress, inflammation, vascular smooth muscle cell apoptosis, and matrix metalloproteinase activation. IMD1-53 inhibited the activation of redox-sensitive signaling pathways, decreased the mRNA and protein expression of nicotinamide adenine dinucleotide phosphate oxidase subunits, and reduced the activity of nicotinamide adenine dinucleotide phosphate oxidase in AAA mice. Expression of Nox4 was upregulated in human AAA segments and in angiotensin II-treated mouse aortas and was markedly decreased by IMD1-53. In vitro, vascular smooth muscle cells with small-interfering RNA knockdown of IMD showed significantly increased angiotensin II-induced reactive oxygen species, and small-interfering RNA knockdown of Nox4 markedly inhibited the reactive oxygen species. IMD knockdown further increased the apoptosis of vascular smooth muscle cells and inflammation, which was reversed by Nox4 knockdown. Preincubation with IMD17-47 and protein kinase A inhibitor H89 inhibited the effect of IMD1-53, reducing Nox4 protein levels. CONCLUSIONS: IMD1-53 could have a protective effect on AAA by inhibiting oxidative stress.


Asunto(s)
Antioxidantes/farmacología , Aorta Abdominal/efectos de los fármacos , Aneurisma de la Aorta Abdominal/prevención & control , Estrés Oxidativo/efectos de los fármacos , Hormonas Peptídicas/farmacología , Adrenomedulina/metabolismo , Angiotensina II , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apoptosis/efectos de los fármacos , Cloruro de Calcio , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dilatación Patológica , Modelos Animales de Enfermedad , Genotipo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , NADPH Oxidasas/metabolismo , Neuropéptidos/metabolismo , Hormonas Peptídicas/metabolismo , Fenotipo , Interferencia de ARN , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección
7.
Clin Exp Hypertens ; 39(5): 460-467, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28534711

RESUMEN

The aims of this study are to observe irisin and urotensin II (UII) levels in serum and placenta in normal pregnant and preeclamptic women and investigate the relationship between expressions irisin and UII, and their association with blood pressure. A total of 67 pregnant subjects were recruited, including 31 healthy and 36 preeclamptic pregnant women. Serum irisin and UII concentrations were measured. Expressions of fibronectin type III domain-containing protein 5 (FNDC5) (irisin precursor) and UII in placenta specimens were performed. There was no significant difference of serum irisin levels between severe preeclamptic (SPE)) patients, mild preeclamptic (MPE) patients and normal controls, while serum UII was significantly higher in preeclamptic women than normal pregnancy. There was no relationship between serum UII and irisin levels. In patients with preeclampsia, serum irisin was negatively associated with systolic and diastolic blood pressure(r = -0.350, P = 0.004, r = -0.307, P = 0.011), while serum UII was positively associated with systolic blood pressure (r = 0.291, P = 0.031). Serum irisin, UII, urinary protein level, BMI and serum creatinine were the independent determinants of blood pressure in preeclampsia by multiple regression analysis. Protein expression of FNDC5 and UII was upregulated in placenta of patients with SPE and positively correlated with systolic blood pressure and urinary protein level. We firstly verify that serum irisin and placental irisin precursor expressions have differently correlated with blood pressure. Expressions of irisin and urotensin II have relationships with blood pressure in patients with preeclampsia.


Asunto(s)
Presión Sanguínea , Fibronectinas/metabolismo , Preeclampsia/metabolismo , Urotensinas/metabolismo , Adulto , Índice de Masa Corporal , Estudios de Casos y Controles , Creatinina/sangre , Diástole , Femenino , Fibronectinas/sangre , Fibronectinas/orina , Humanos , Hipertensión/metabolismo , Placenta/metabolismo , Preeclampsia/sangre , Preeclampsia/orina , Embarazo , Proteinuria/orina , Índice de Severidad de la Enfermedad , Sístole , Urotensinas/sangre , Urotensinas/orina
8.
Sheng Li Ke Xue Jin Zhan ; 48(1): 4-11, 2017 Feb.
Artículo en Zh | MEDLINE | ID: mdl-29927207

RESUMEN

The sulfur-containing gases hydrogen sulfide (H2S)and sulfur dioxide (SO2 )were previously considered to be waste gases. Recent studies showed that they could be endogenously generated from metabolism of the sulfur-containing amino acids in mammals. Endogenous H2S and SO2 generation pathways also existed in the cardiovascular system.H2S and SO2 had important physiological effects in the cardio-vascular system including vasorelaxation and myocardial negative inotropic effect. The pathophysiological effects of H2S and SO2 in the cardiovascular system have been recognized, such as alleviating hypertension and pulmonary hypertension, inhibiting the development of atherosclerosis, and protecting against myocardial ischemia-reperfusion (I /R)injury and isoproterenol-induced myocardial injury. Adenosine triphosphate-sensitive potassium (KATP )channel, L-type calcium (L-Ca2 +) channel, cGMP, NF-κB signaling pathway and MAPK signaling pathway and so on participated in the biological effects of H2S and SO2 .The above findings suggested that H2S and SO2 were important endogenous gaseous signaling molecules in the cardiovascular system, which provided a new way to elucidate the pathogenesis and therapeutic targets of cardiovascular diseases.


Asunto(s)
Sistema Cardiovascular/metabolismo , Sulfuro de Hidrógeno/metabolismo , Transducción de Señal , Dióxido de Azufre/metabolismo , Animales , Aterosclerosis , Canales de Calcio Tipo L/metabolismo , Hipertensión Pulmonar , Daño por Reperfusión Miocárdica , Canales de Potasio/metabolismo , Azufre
9.
Kidney Int ; 89(3): 586-600, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26880455

RESUMEN

Deficiency in α-Klotho is involved in the pathogenesis of vascular calcification. Since intermedin (IMD)1-53 (a calcitonin/calcitonin gene-related peptide) protects against vascular calcification, we studied whether IMD1-53 inhibits vascular calcification by upregulating α-Klotho. A rat model of chronic kidney disease (CKD) with vascular calcification induced by the 5/6 nephrectomy plus vitamin D3 was used for study. The aortas of rats with CKD showed reduced IMD content but an increase of its receptor, calcitonin receptor-like receptor, and its receptor modifier, receptor activity-modifying protein 3. IMD1-53 treatment reduced vascular calcification. The expression of α-Klotho was greatly decreased in the aortas of rats with CKD but increased in the aortas of IMD1-53-treated rats with CKD. In vitro, IMD1-53 increased α-Klotho protein level in calcified vascular smooth muscle cells. α-Klotho knockdown blocked the inhibitory effect of IMD1-53 on vascular smooth muscle cell calcification and their transformation into osteoblast-like cells. The effect of IMD1-53 to upregulate α-Klotho and inhibit vascular smooth muscle cell calcification was abolished by knockdown of its receptor or its modifier protein, or treatment with the protein kinase A inhibitor H89. Thus, IMD1-53 may attenuate vascular calcification by upregulating α-Klotho via the calcitonin receptor/modifying protein complex and protein kinase A signaling.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Glucuronidasa/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Hormonas Peptídicas/farmacología , Insuficiencia Renal Crónica/tratamiento farmacológico , Calcificación Vascular/prevención & control , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Aorta Torácica/patología , Células Cultivadas , Colecalciferol , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Glucuronidasa/genética , Humanos , Proteínas Klotho , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Nefrectomía , Osteoblastos/metabolismo , Osteoblastos/patología , Fenotipo , Interferencia de ARN , Ratas Sprague-Dawley , Proteína 3 Modificadora de la Actividad de Receptores/metabolismo , Receptores de Calcitonina/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Transducción de Señal/efectos de los fármacos , Transfección , Regulación hacia Arriba , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
10.
Kidney Blood Press Res ; 41(4): 434-49, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27467277

RESUMEN

BACKGROUND/AIMS: Urotensin II (UII) and its receptor are highly expressed in the kidney tissue of patients with diabetic nephropathy (DN). The aim of this study is to examine the roles of UII in the induction of endoplasmic reticulum stress (ER stress) and Epithelial-mesenchymal transition (EMT) in DN in vivo and in vitro. METHODS: Kidney tissues were collected from patients with DN. C57BL/6 mice and mice with UII receptor knock out were injected with two consecutive doses of streptozotocin to induce diabetes and were sacrificed at 3th week for in vivo study. HK-2 cells in vitro were cultured and treated with UII. Markers of ER stress and EMT, fibronectin and type IV collagen were detected by immunohistochemistry, real time PCR and western blot. RESULTS: We found that the expressions of protein of UII, GRP78, CHOP, ALPHA-SMA, fibronectin and type IV collagen were upregulated while E-cadherin protein was downregulated as shown by immunohistochemistry or western blot analysis in kidney of diabetic mice in comparison to normal control; moreover expressions of GRP78, CHOP, ALPHA-SMA, fibronectin and type IV collagen were inhibited while E-caherin expression was enhanced in kidney in diabetic mice with UII receptor knock out in comparison to C57BL/6 diabetic mice. In HK-2 cells, UII induced upregulation of GRP78, CHOP, ALPHA-SMA, fibroblast-specifc protein 1(FSP-1), fibronectin and type collagen and downregulation of E-cadherin. UII receptor antagonist can block UII-induced ER stress and EMT; moreover, 4-PBA can inhibit the mRNA expression of ALPHA-SMA and FSP1 induced by UII in HK-2 cells. CONCLUSIONS: We are the first to verify UII induces ER stress and EMT and increase extracellular matrix production in renal tubular epithelial cell in early diabetic mice. Moreover, UII may induce renal tubular epithelial EMT via triggering ER stress pathway in vitro, which might be the new pathogenic pathway for the development of renal fibrosis in DN.


Asunto(s)
Nefropatías Diabéticas/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Túbulos Renales/patología , Urotensinas/farmacología , Animales , Línea Celular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/metabolismo , Chaperón BiP del Retículo Endoplásmico , Células Epiteliales/metabolismo , Células Epiteliales/patología , Matriz Extracelular/metabolismo , Humanos , Túbulos Renales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Kidney Blood Press Res ; 41(1): 78-85, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26870964

RESUMEN

AIMS/INTRODUCTION: Irisin is a newly identified myokine which can promote energy expenditure. Urotensin II (UII) is identified as the most potent mammalian vasoconstrictor to date. Previous studies showed that UII can aggravate insulin resistance while irisin alleviate insulin resistance. Through this study, it is our aim to elucidate if UII can induce insulin resistance and also have an association with the irisin level in hemodialysis (HD) patients. MATERIALS AND METHODS: One hundred and twenty-eight patients on maintenance hemodialysis treatment and forty healthy subjects were enrolled in this study. Blood irisin concentrations and UII concentrations were measured by ELISA and RIA respectively. The body composition was analyzed by bioelectrical impedance. RESULTS: The serum irisin levels and UII levels were both significantly lower in HD patients in comparison to that of the healthy subjects. The serum irisin levels were lower in HD patients with protein energy wasting than those of the patients without protein energy wasting. The independent determinants of circulating Ln (irisin) (the natural logarithm of irisin) were UII lean body mass and patients with protein energy wasting. CONCLUSIONS: Our results are the first to provide the clinical evidence of the association among irisin, UII, and protein energy wasting. Our results hint that UII and protein energy wasting might inhibit the release or synthesis of irisin from skeletal muscles in HD patients.


Asunto(s)
Fibronectinas/sangre , Desnutrición Proteico-Calórica/sangre , Desnutrición Proteico-Calórica/diagnóstico , Diálisis Renal , Urotensinas/sangre , Adulto , Anciano , Biomarcadores/sangre , Composición Corporal/fisiología , Ejercicio Físico/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Diálisis Renal/tendencias
12.
Apoptosis ; 20(4): 512-22, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25608996

RESUMEN

NOD1 is a member of nucleotide-binding oligomerization domain-like receptors family that participates in many inflammatory processes. Previous studies demonstrated that NOD1 plays an important role in inflammatory cardiovascular diseases. However, its role in myocardial ischemia/reperfusion (I/R) injury remains unknown. The present study investigate whether NOD1 is involved in the pathogenesis of mouse myocardial I/R injury and the underlying mechanisms. Administration of NOD1 ligand (DAP) significantly enhanced myocardial I/R injury, as demonstrated by increased infarct size, the number of TUNEL-positive nuclei, caspase-3 activity, the infiltration of Mac-2- and IL-6-positive cells as compared with untreated heart or cardiomyocytes after I/R injury. In contrast, knockdown of NOD1 by siRNA markedly attenuated mimetic I/R induced cardiomyocyte apoptosis in vitro, indicating that NOD1 enhanced myocardial I/R injury partially through direct heart effects. These effects were partially associated with activation of JNK, p38 MAPK and NF-κB signaling pathways. Taken together, these results provide the first evidence that activation of intracellular sensor NOD1 enhances myocardial I/R injury and may provide novel therapeutic target for ameliorating the ischemic heart diseases.


Asunto(s)
Ácido Diaminopimélico/metabolismo , Isquemia Miocárdica/cirugía , Daño por Reperfusión Miocárdica/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Transducción de Señal , Animales , Apoptosis , Humanos , Ligandos , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/genética , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/genética
13.
Mol Cell Biochem ; 392(1-2): 65-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24710639

RESUMEN

Vascular calcification (VC) is highly associated with increased morbidity and mortality in patients with advanced chronic kidney disease. Paracrine/autocrine factors such as vasoactive peptides are involved in VC development. Here, we investigated the expression of the novel peptide C-type natriuretic peptide (CNP) in the vasculature, tested its ability to prevent VC in vivo and in vitro, and examined the mechanism involved. Rat aortic VC was induced by vitamin D3 plus nicotine (VDN). CNP (500 ng/kg/h) was administered by mini-osmotic pump. Calcification was examined by von Kossa staining; CNP and cyclic guanosine monophosphate (cGMP) contents were detected by radioimmunoassay, and mRNA and protein levels were examined by real-time PCR and Western blot analysis in aortas and calcified vascular smooth muscle cells (VSMCs). VDN-treated rat aortas showed higher CNP content and decreased expression of its receptor natriuretic peptide receptor B, along with increased vascular calcium deposition and alkaline phosphatase (ALP) activity. Low CNP levels were accompanied by increased vascular calcium deposition and ALP activity in VDN-treated rats when compared to vehicle treatment, which was further confirmed in cultured VSMCs. Administration of CNP greatly reduced VC in VDN-treated aortas compared with controls, which was confirmed in calcified VSMCs. The decrease in alpha-actin expression was ameliorated by CNP in vitro. Moreover, protein expression levels of osteopontin (OPN) were significantly up-regulated in calcified aortas, and CNP increased OPN expression in calcified aortas. Furthermore, CNP downregulated OPN and bone morphogenic protein 2 (BMP-2) expression in calcified aortas and VSMCs. Modulation of OPN and BMP-2 expression by CNP and the beneficial effects of CNP on calcified VSMCs were blocked significantly by protein kinase G inhibitor H7. Impaired local endogenous CNP and its receptor system may be associated with increased mineralization in vivo in rat aortas with VC, and administration of CNP inhibits VC development in vivo and in vitro, at least in part, via a cGMP/PKG pathway.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Proteína Morfogenética Ósea 2/metabolismo , Calcinosis/prevención & control , Péptido Natriurético Tipo-C/farmacología , Osteopontina/metabolismo , Animales , Secuencia de Bases , Vasos Sanguíneos/patología , Cartilla de ADN , Masculino , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
14.
Apoptosis ; 18(9): 1132-44, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23686245

RESUMEN

Our previous work reported that endoplasmic reticulum stress (ERS)-mediated apoptosis was activated during vascular calcification (VC). Activating transcription factor 4 (ATF4) is a critical transcription factor in osteoblastogenesis and ERS-induced apoptosis. However, whether ATF4 is involved in ERS-mediated apoptosis contributing to VC remains unclear. In the present study, in vivo VC was induced in rats by administering vitamin D3 plus nicotine. Vascular smooth muscle cell (VSMC) calcification in vitro was induced by incubation in calcifying media containing ß-glycerophosphate and CaCl2. ERS inhibitors taurine or 4-phenylbutyric acid attenuated ERS and VSMC apoptosis in calcified rat arteries, reduced calcification and retarded the VSMC contractile phenotype transforming into an osteoblast-like phenotype in vivo. Inhibition of ERS retarded the VSMC phenotypic transition into an osteoblast-like cell phenotype and reduced VSMC calcification and apoptosis in vitro. Interestingly, ATF4 was activated in calcified aortas and calcified VSMCs in vitro. ATF4 knockdown attenuated ERS-induced apoptosis in calcified VSMCs. ATF4 deficiency blocked VSMC calcification and negatively regulated the osteoblast phenotypic transition of VSMCs in vitro. Our results demonstrate that ATF4 was involved at least in part in the process of ERS-mediated apoptosis contributing to VC.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Apoptosis , Estrés del Retículo Endoplásmico , Músculo Liso Vascular/citología , Calcificación Vascular/metabolismo , Calcificación Vascular/fisiopatología , Factor de Transcripción Activador 4/genética , Animales , Células Cultivadas , Humanos , Masculino , Músculo Liso Vascular/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Ratas , Ratas Sprague-Dawley , Calcificación Vascular/genética
15.
Nitric Oxide ; 32: 56-61, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23629152

RESUMEN

BACKGROUND: The study was designed to explore if sulfur dioxide (SO2) preconditioning increased antioxidative capacity in rat with myocardial ischemia reperfusion (I/R) injury. METHODS: The myocardial I/R model was made by left coronary artery ligation for 30min and reperfusion for 120min in rats. Myocardial infarct size and plasma lactate dehydrogenase (LDH) and creatine kinase (CK) activities, plasma superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-Px) and glutathione (GSH) changes were detected for the rats. The contents of myocardial hydrogen sulfide (H2S) and nitric oxide (NO) were measured. Myocardial protein expressions of SOD1, SOD2, cystathionine γ-lyase (CSE) and iNOS were tested using Western blot. RESULTS: Myocardial infarction developed and plasma CK and LDH activities were significantly increased in I/R group compared with those in control group, but SO2 preconditioning significantly reduced myocardial infarct size, and plasma CK and LDH activities. SO2 preconditioning successfully increased plasma SOD, GSH and GSH-Px levels and myocardial SOD1 protein expression, but decreased MDA level in rats of I/R group. Compared with controls, the myocardial H2S level and CSE expression were decreased after I/R, but myocardial NO level and iNOS expression were increased. With the treatment of SO2, myocardial H2S level and CSE expression were increased, but myocardial NO level and iNOS expression were decreased compared with those in I/R group. CONCLUSIONS: SO2 preconditioning could significantly reduce I/R-induced myocardial injury in vivo in association with increased myocardial antioxidative capacity, upregulated myocardial H2S/CSE pathway but downregulated NO/iNOS pathway.


Asunto(s)
Antioxidantes/metabolismo , Precondicionamiento Isquémico Miocárdico/métodos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Dióxido de Azufre/farmacología , Animales , Creatina Quinasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Glutatión/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Masculino , Infarto del Miocardio/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
16.
Acta Pharmacol Sin ; 34(4): 501-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23524571

RESUMEN

AIM: To explore the mechanisms underlying the protection by SO2 preconditioning against rat myocardial ischemia/reperfusion (I/R) injury. METHODS: Male Wistar rats underwent 30-min left coronary artery ligation followed by 120-min reperfusion. An SO2 donor (1 µmol/kg) was intravenously injected 10 min before the ischemia, while LY294002 (0.3 mg/kg) was intravenously injected 30 min before the ischemia. Plasma activities of LDH and CK were measured with an automatic enzyme analyzer. Myocardial infarct size was detected using Evans-TTC method. The activities of caspase-3 and -9 in myocardium were assayed using a commercial kit, and the levels of p-Akt, Akt, PI3K and p-PI3K were examined with Western blotting. RESULTS: Pretreatment with SO2 significantly reduced the myocardial infarct size and plasma LDH and CK activities, as well as myocardial caspase-3 and -9 activities in the rats. Furthermore, the pretreatment significantly increased the expression levels of myocardial p-Akt and p-PI3K p85. Administration of the PI3K inhibitor LY294002 blocked all the effects induced by SO2 pretreatment. CONCLUSION: The results suggest that the PI3K/Akt pathway mediates the protective effects of SO2 preconditioning against myocardial I/R injury in rats.


Asunto(s)
Precondicionamiento Isquémico Miocárdico/métodos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Dióxido de Azufre/farmacología , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Cromonas/farmacología , Vasos Coronarios/metabolismo , Masculino , Morfolinas/farmacología , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal
17.
Beijing Da Xue Xue Bao Yi Xue Ban ; 45(3): 489-92, 2013 Jun 18.
Artículo en Zh | MEDLINE | ID: mdl-23774934

RESUMEN

OBJECTIVE: To establish a simple method of measuring hydrogen sulfide (H2S) in cultured living cells. METHODS: Filtration membrane was stuck on the lid of cell culture plate. H2S released from cultured cells was trapped by zinc acetate to generate ZnS deposition. Then the ZnS trapped in the filtration membrane was measured by methylene blue assay and the H2S production from the living cells was counted according to the standard curve. This simple method was used to access the H2S release in HepG2 (high expression CBS and CSE) and HUVEC (low expression CSE) cell lines. RESULTS: H2S generation in cultured HepG2 cells assayed using the present method was (859.39±19.12) nmol/(min×10(6) cells). PAG (CSE inhibitor), HA (CBS inhibitor) or the two-inhibitor (PAG+HA) treatment significantly lowered H2S release, respectively: (341.34±105.90) nmol/(min×10(6) cells), (375.05±174.50) nmol/(min×10(6) cells), and (204.47±97.14) nmol/(min×10(6) cells). The H2S production of HUVEC was (26.23±3.24) nmol/(min×10(6) cells) (about 1/30 production of HepG2 cell). Trypan blue assay showed that the cell viability was greater than 95%, suggesting that there was no cytotoxicity by using the present instrument. CONCLUSION: The modified instrument in cell culture plate lid was feasible for detection of hydrogen sulfide release in living cells.


Asunto(s)
Sulfuro de Hidrógeno/análisis , Supervivencia Celular , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos
18.
Beijing Da Xue Xue Bao Yi Xue Ban ; 45(2): 192-6, 2013 Apr 18.
Artículo en Zh | MEDLINE | ID: mdl-23591335

RESUMEN

OBJECTIVE: To investigate the role of hydrogen sulfide (H2S) on oxidized-low density lipoprotein (ox-LDL)-stimulated NF-κB pathway in human monocytes/macrophages and its mechanisms. METHODS: THP-1 cells were induced to differentiate into macrophages by incubation with phorbol myristate acetate (PMA) . The human monocytes/macrophages were divided into 4 groups: control group, ox-LDL group, ox-LDL + H2S 100 µmol/L group and ox-LDL + H2S 500 µmol/L group. The expression of IκBα and phosphorylation of NF-κB p65 in the cells were detected by Western blotting. The expression of IκBα and nuclear translocation of NF-κB p65 in the cells were observed by laser confocal method. The interaction between NF-κB p65 and IκBα in the nuclear extracts was detected by coimmunoprecipitation method. RESULTS: Compared with the control group, the phosphorylation of NF-κB p65 in the human monocytes/macrophages of ox-LDL group was increased significantly (0.855 ± 0.116 vs. 0.502 ± 0.218, P=0.046), while the expression of IκBα in the cells of the ox-LDL group was decreased (0.612 ± 0.216 vs. 0.997 ± 0.167, P=0.029). Compared with the ox-LDL group, the phosphorylation of NF-κB p65 in the cells of the ox-LDL + H2S 100 µmol/L group and the ox-LDL + H2S 500 µmol/L group was decreased significantly (0.424 ± 0.225 vs. 0.855 ± 0.116, P=0.020; 0.378 ± 0.071 vs. 0.855 ± 0.116, P=0.011, respectively), while the expressions of IκBα in the cells of the ox-LDL + H2S 100 µmol/L group and the ox-LDL + H2S 500 µmol/L group were increased (1.037 ± 0.111 vs. 0.612 ± 0.216, P=0.015; 1.046 ± 0.084 vs. 0.612 ± 0.216, P=0.013, respectively). The results from laser confocal method demonstrated that the IκBα expression in the cytoplasma of cells in the ox-LDL group was lower than that in the control group, and the nuclear translocation of NF-κB p65 in the cells of the ox-LDL group was higher than that in the control group. The IκBα expression in the cytoplasma of cells in the ox-LDL+ H2S 100 µmol/L group and ox-LDL + H2S 500 µmol/L group was higher than that in the ox-LDL group, and the nuclear translocation of NF-κB p65 in the cells of ox-LDL group was lower than that in the ox-LDL group. The coimmunoprecipitation experiment showed that no IκBα integrated with NF-κB p65 was detected in the nuclear extracts of cells in the control group, a small amount of IκBα integrated with NF-κB p65 was detected in the nuclear extracts of cells in the ox-LDL group, but a large amount of IκBα integrated with NF-κB p65 was detected in the nuclear extracts of cells in the ox-LDL+H2S 100 µmol/L group and the ox-LDL + H2S 500 µmol/L group. CONCLUSION: H2S inhibited the activation of NF-κB p65 pathway in the ox-LDL-induced human monocytes/macrophages. The mechanisms might involve the prevention of the degradation of IκBα, then the inhibition of the phosphorylation and nuclear translocation of NF-κB p65, thus promoting the IκBα integrated with NF-κB p65 in the nuclei, and then inhibiting the activity of NF-κB.


Asunto(s)
Sulfuro de Hidrógeno/farmacología , Lipoproteínas LDL/farmacología , Macrófagos/metabolismo , Monocitos/metabolismo , Factor de Transcripción ReIA/metabolismo , Línea Celular , Humanos , Proteínas I-kappa B/metabolismo , Inhibidor NF-kappaB alfa , Transducción de Señal/efectos de los fármacos
19.
Sheng Li Ke Xue Jin Zhan ; 44(2): 81-6, 2013 Apr.
Artículo en Zh | MEDLINE | ID: mdl-23847916

RESUMEN

Leptin is a product of the obese (ob) gene and acts through its receptor Ob-R. Leptin is primarily known for its role as a hypothalamic modulator of food, especially in intake, energy balance, fat stores and body weight. Recent studies have shown that leptin may be involved in the development of respiratory diseases such as pulmonary artery hypertension, chronic obstructive pulmonary disease, lung neoplasms and asthma. Therefore, further studies are needed to elucidate the mechanisms accounting for the association between leptin and respiratory diseases, which may lead to the development of novel approaches to the prevention and treatment of these diseases. Here we give an overview of the distribution and physiological function of leptin and ob-R, and summarize the recent progress in the relationship between leptin and respiratory diseases.


Asunto(s)
Leptina/fisiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Enfermedades Respiratorias/fisiopatología , Animales , Asma/fisiopatología , Humanos , Leptina/genética , Neoplasias Pulmonares/fisiopatología , Receptores de Leptina/fisiología
20.
World J Pediatr ; 19(4): 390-400, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36781629

RESUMEN

BACKGROUND: The present work was designed to explore whether electrocardiogram (ECG) index-based models could predict the effectiveness of metoprolol therapy in pediatric patients with postural tachycardia syndrome (POTS). METHODS: This study consisted of a training set and an external validation set. Children and adolescents with POTS who were given metoprolol treatment were enrolled, and after follow-up, they were grouped into non-responders and responders depending on the efficacy of metoprolol. The difference in pre-treatment baseline ECG indicators was analyzed between the two groups in the training set. Binary logistic regression analysis was further conducted on the association between significantly different baseline variables and therapeutic efficacy. Nomogram models were established to predict therapeutic response to metoprolol. The receiver-operating characteristic curve (ROC), calibration, and internal validation were used to evaluate the prediction model. The predictive ability of the model was validated in the external validation set. RESULTS: Of the 95 enrolled patients, 65 responded to metoprolol treatment, and 30 failed to respond. In the responders, the maximum value of the P wave after correction (Pcmax), P wave dispersion (Pd), Pd after correction (Pcd), QT interval dispersion (QTd), QTd after correction (QTcd), maximum T-peak-to-T-end interval (Tpemax), and T-peak-to-T-end interval dispersion (Tped) were prolonged (all P < 0.01), and the P wave amplitude was increased (P < 0.05) compared with those of the non-responders. In contrast, the minimum value of the P wave duration after correction (Pcmin), the minimum value of the QT interval after correction (QTcmin), and the minimum T-peak-to-T-end interval (Tpemin) in the responders were shorter (P < 0.01, < 0.01 and < 0.01, respectively) than those in the non-responders. The above indicators were screened based on the clinical significance and multicollinearity analysis to construct a binary logistic regression. As a result, pre-treatment Pcmax, QTcmin, and Tped were identified as significantly associated factors that could be combined to provide an accurate prediction of the therapeutic response to metoprolol among the study subjects, yielding good discrimination [area under curve (AUC) = 0.970, 95% confidence interval (CI) 0.942-0.998] with a predictive sensitivity of 93.8%, specificity of 90.0%, good calibration, and corrected C-index of 0.961. In addition, the calibration curve and standard curve had a good fit. The accuracy of internal validation with bootstrap repeated sampling was 0.902. In contrast, the kappa value was 0.769, indicating satisfactory agreement between the predictive model and the results from the actual observations. In the external validation set, the AUC for the prediction model was 0.895, and the sensitivity and specificity were 90.9% and 95.0%, respectively. CONCLUSIONS: A high-precision predictive model was successfully developed and externally validated. It had an excellent predictive value of the therapeutic effect of metoprolol on POTS among children and adolescents.


Asunto(s)
Metoprolol , Síndrome de Taquicardia Postural Ortostática , Humanos , Niño , Adolescente , Metoprolol/uso terapéutico , Síndrome de Taquicardia Postural Ortostática/diagnóstico , Síndrome de Taquicardia Postural Ortostática/tratamiento farmacológico , Frecuencia Cardíaca , Sensibilidad y Especificidad , Curva ROC
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA