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.
Int J Mol Med ; 51(3)2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36704846

RESUMEN

Filamin A (FLNA) is a high molecular weight cytoskeleton protein important for cell locomotion. A relationship between FLNA mutations and pulmonary arterial hypertension (PAH) has previously been reported; however, the detailed mechanism remains unclear. The present study aimed to explore the role of FLNA in vascular smooth muscle cells during the development of PAH. Smooth muscle cell (SMC)­specific FLNA­deficient mice were generated and the mice were then exposed to hypoxia for 28 days to build the mouse model of PAH. Human pulmonary arterial smooth muscle cells (PASMCs) were also cultured and transfected with FLNA small interfering RNA or overexpression plasmids to investigate the effects of FLNA on PASMC proliferation and migration. Notably, compared with control individuals, the expression levels of FLNA were increased in lung tissues from patients with PAH, and it was obviously expressed in the PASMCs of pulmonary arterioles. FLNA deficiency in SMCs attenuated hypoxia­induced pulmonary hypertension and pulmonary vascular remodeling. In vitro studies suggested that absence of FLNA impaired PASMC proliferation and migration, and produced lower levels of phosphorylated (p)­PAK­1 and RAC1 activity. However, FLNA overexpression promoted PASMC proliferation and migration, and increased the expression levels of p­PAK­1 and RAC1 activity. The present study highlights the role of FLNA in pulmonary vascular remodeling; therefore, it could serve as a potential target for the treatment of PAH.


Asunto(s)
Filaminas , Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Animales , Humanos , Ratones , Proliferación Celular , Células Cultivadas , Filaminas/genética , Filaminas/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipoxia/complicaciones , Hipoxia/genética , Hipoxia/metabolismo , Miocitos del Músculo Liso/metabolismo , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/metabolismo , Arteria Pulmonar/metabolismo , Transducción de Señal , Remodelación Vascular/genética
2.
Biomed Res Int ; 2022: 9294148, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36246958

RESUMEN

Right ventricular (RV) failure determines the prognosis in pulmonary arterial hypertension (PAH), but the underlying mechanism is still unclear. Growing evidence has shown that microRNAs participate in RV remodeling. This study is undertaken to explore the role of miR-335-5p in regulating RV remodeling induced by PAH. Two PAH models were used in the study, including the monocrotaline rat model and hypoxia/su5416 mouse model. miRNA sequencing and RT-qPCR validation identified that miR-335-5p was elevated in the RV of PAH rats. In vitro, miR-335-5p expression was increased after angiotensin II treatment, and miR-335-5p inhibition relieved angiotensin II-induced cardiomyocyte hypertrophy. The luciferase reporter assay showed that calumenin was a target gene for miR-335-5p. Pretreatment with miR-335-5p inhibitors could rescue calumenin downregulation induced by angiotensin II in H9C2 cells. Moreover, intracellular Ca2+ concentration and apoptosis were increased after angiotensin II treatment, and miR-335-5p inhibition decreased intracellular Ca2+ accumulation and apoptosis. Finally, in vivo miR-335-5p downregulation (antagomir miR-335-5p) attenuated RV remodeling and rescued calumenin downregulation under conditions of hypoxia/su5416 exposure. Our work highlights the role of miR-335-5p and calumenin in RV remodeling and may lead to the development of novel therapeutic strategies for right heart failure.


Asunto(s)
Hipertensión Pulmonar , MicroARNs , Hipertensión Arterial Pulmonar , Angiotensina II/metabolismo , Animales , Antagomirs , Hipertensión Pulmonar Primaria Familiar , Hipertensión Pulmonar/metabolismo , Hipoxia/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Monocrotalina , Hipertensión Arterial Pulmonar/genética , Ratas , Regulación hacia Arriba/genética , Remodelación Ventricular/genética
3.
Pulm Circ ; 12(3): e12096, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35911182

RESUMEN

Pulmonary hypertension secondary to heart failure (HF-PH) combined with pulmonary vascular remodeling has a high mortality rate. Apolipoprotein A1 (ApoA1) has been shown to adversely affect outcomes in patients with HF. A prospective follow-up study was performed on 239 consecutive patients with HF-PH who underwent right heart catheterization. Proteomics technology was used to analyze different proteins in plasma between post- and precapillary pulmonary hypertension (CpcPH) and isolated postcapillary pulmonary hypertension (IpcPH) filtered by propensity score matching. Ultimately, 175 patients were enrolled and followed for an average of 4.4 years. Lipoprotein components in plasma were measured, and the following clinical events were tracked. Proteomics data showed that lipid metabolism and inflammation were different between CpcPH and IpcPH. ApoA1 levels in HF-PH patients with CpcPH were lower than those in HF-PH patients with IpcPH. The patients with lower ApoA1 levels (≤1.025 g/L) were in a higher New York Heart Association class and had high levels of NT-proBNP, mean pulmonary artery pressure, PVR, and diastolic pressure gradient. Besides, HF-PH patients with lower ApoA1 levels had a 2.836-fold higher relative risk of comorbid CpcPH compared with patients with higher ApoA1 levels. Moreover, patients with lower ApoA1 levels had a lower survival rate after adjusting for CpcPH. In conclusion, ApoA1 levels were negatively correlated with PVR levels. Lower ApoA1 levels were an independent risk factor for pulmonary vascular remodeling in HF-PH patients. The survival of HF-PH patients with lower ApoA1 levels was reduced.

4.
Environ Toxicol ; 37(4): 730-740, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34921586

RESUMEN

The clinical application of doxorubicin (Dox) is limited due to its cardiotoxicity, while the pathogenesis remains to be fully understood. Recent studies have suggested that microRNA (miRNA) plays an important role in Dox-induced cardiotoxicity. This work aims to investigate the effects of miR-125b in Dox-induced cardiotoxicity. Here, mice model combined with cell line analysis were used, and cell viability assay, detection of reactive oxygen species (ROS), malondialdehyde (MDA) activity, lactate dehydrogenase (LDH) activity, glutathione (GSH) level, glutathione peroxidase (GSH-Px) level, superoxide dismutase (SOD) activity, and histopathological changes were performed to characterize miR-125b effects; real-time quantitative polymerase chain reaction (PCR), luciferase reporter assay, RNA immunoprecipitation, and western blot analysis were subjected to reveal the underlying mechanisms. It was found that miR-125b level was upregulated in myocardial cell line H9C2 treated with Dox and miR-125b overexpression enhanced Dox-induced cytotoxicology of H9C2 cells, while miR-125b inhibition exhibited a protective effect by measuring ROS level and cell viability. In consistent, in vivo experiments with miR-125b agomir or antagomir obtained a consistent result through examining the activity of MDA, LDH, GSH, GSH-Px, SOD, and histopathological changes. Furthermore, we found that miR-125b could target STARD13 and thus suppressed the nucleus-cytoplasmic translocation of yes-associated protein (YAP). Additionally, this STARD13/YAP axis is necessary for miR-125b-mediated regulation on Dox-induced cytotoxicology of H9C2 cells. In conclusion, our study demonstrated that miR-125b could enhance Dox-induced cardiotoxicity through targeting the STARD13/YAP axis.


Asunto(s)
Cardiotoxicidad , MicroARNs , Animales , Cardiotoxicidad/etiología , Doxorrubicina/toxicidad , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos , Especies Reactivas de Oxígeno/metabolismo
5.
Exp Cell Res ; 402(1): 112552, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33711329

RESUMEN

Endothelial dysfunction is an early step in the development of atherosclerotic cardiovascular disease. Iron overload can lead to excessive mitochondrial reactive oxygen species (mtROS) production, resulting in mitochondrial dysfunction and vascular endothelial cell (EC) damage. Mitoferrin 2 (Mfrn2) is an iron transporter in the inner mitochondrial membrane. This study aimed to assess whether Mfrn2 and mitochondrial iron overload were involved in atherosclerosis progression and to explore the potential mechanism. We observed significant upregulation of Mfrn2 in the arteries of high-fat diet (HFD)-fed Apolipoprotein E-/- (ApoE-/-) mice and in TNF-α-induced mouse aortic endothelial cells (MAECs). Mfrn2 gene silencing inhibited mitochondrial iron overload, stabilized mitochondrial membrane potential and improved mitochondrial function in TNF-α-induced MAECs. Vascular EC-specific knockdown of Mfrn2 in ApoE-/- mice markedly decreased atherosclerotic lesion formation and the levels of ICAM-1 in aortas and reduced monocyte infiltration into the vascular wall. Furthermore, TNF-α increased the binding of 14-3-3 epsilon (ε) and Mfrn2, preventing Mfrn2 degradation and leading to mitochondrial iron overload in ECs, while 14-3-3ε overexpression increased Mfrn2 stability by inhibiting its ubiquitination. Together, our results reveal that Mfrn2 deficiency attenuates endothelial dysfunction by decreasing iron levels within the mitochondria and mitochondrial dysfunction. These findings may provide new insights into preventive and therapeutic strategies against vascular endothelial dysfunction in atherosclerotic disease.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/genética , Proteínas de Transporte de Catión/genética , Sobrecarga de Hierro/genética , Mitocondrias/metabolismo , Proteínas 14-3-3/genética , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Endotelio/lesiones , Endotelio/metabolismo , Endotelio/patología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/metabolismo , Ratones Noqueados , Mitocondrias/patología , Factor de Necrosis Tumoral alfa/genética
6.
Cell Signal ; 80: 109900, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33370582

RESUMEN

Low shear stress (LSS) plays a critical role in the development of atherosclerotic plaques and vascular inflammation. Previous studies have reported Akt phosphorylation induced by LSS. However, the mechanism and role of Akt activation remains unclear in LSS-induced endothelial dysfunction. In this study, our results demonstrated the increased phosphorylation of IKKε, TBK1 and Akt in HUVECs exposed to LSS. Furthermore, IKKε silencing using small interfering RNAs significantly reduced LSS-induced Akt phosphorylation. In contrast, silencing of TBK1 or inhibition of PI3K and mTORC2 had no effect on LSS-induced Akt phosphorylation. Notably, Akt inhibition markedly diminished LSS-induced expression of ICAM-1, VCAM-1 and MCP-1, as well as LSS-induced IRF3 phosphorylation and nuclear translocation, without affecting the activation of NF-κB and STAT1. Moreover, endothelial cell specific Akt overexpression mediated by adeno-associated virus markedly increased intimal ICAM-1 and IRF3 expression at LSS area of partially ligated carotid artery in mice. In brief, our findings suggest that LSS-induced Akt phosphorylation is positively regulated by IKKε and promotes IRF3 activation, leading to endothelial inflammation.


Asunto(s)
Quinasa I-kappa B/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Resistencia al Corte , Animales , Aorta/metabolismo , Aorta/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Inflamación/patología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT1/metabolismo
7.
Cell Tissue Res ; 382(3): 585-598, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32719938

RESUMEN

Pyruvate kinase M2 (PKM2), which is encoded by PKM, is a ubiquitously expressed intracellular protein and is associated with proliferation cell phenotype. In PAH patients and PAH models, we found higher levels of PKM2 tyrosine 105 phosphorylation (phospho-PKM2 (Y105)) than in controls, both in vivo and in vitro. Here, we demonstrate that PKM2 stimulates inflammatory and apoptosis signalling pathways in pulmonary artery smooth muscle cells (PASMCs) and promotes PASMC migration and proliferation. PKM2 phosphorylation promoted the dimerization activation and nuclear translocation of STAT3, a transcription factor regulating proliferation, growth, and apoptosis. TLR2, a transmembrane protein receptor involved in both innate and adaptive immune responses, promoted PKM2 phosphorylation in hypoxia-induced PASMCs. Therefore, we hypothesized that PKM2 also affects the proliferation and migration of PASMCs. The proliferation of hypoxia-induced normal human pulmonary artery smooth muscle cells (normal-HPASMCs) was found to be inhibited by TEPP-46 (PKM2 agonist) and PKM2 siRNA using wound healing, 5-ethynyl-2'-deoxyuridine (EdU), and immunofluorescence (Ki67) assays. PASMCs isolated from PAH patients (PAH-HPASMCs) and hypoxia-treated rats (PAH-RPASMCs) also confirmed the above results. TEPP-46 treatment was found to improve hypoxia-induced pulmonary artery remodelling and right heart function in mice, and the link between PKM2 and STAT3 was also confirmed in vivo. In conclusion, PKM2 plays crucial roles in the proliferation and migration of PASMCs.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Miocitos del Músculo Liso/metabolismo , Piruvato Quinasa/metabolismo , Remodelación Vascular/fisiología , Animales , Movimiento Celular , Proliferación Celular , Humanos , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/patología , Masculino , Ratones , Transfección
8.
Exp Cell Res ; 380(1): 36-46, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30802452

RESUMEN

Pulmonary arterial hypertension (PAH) is a diffuse pulmonary microvascular remodeling disease accompanied by malignant proliferation of pulmonary artery smooth muscle cells (PASMCs), which causes persistent pulmonary artery pressure elevation, right ventricular hypertrophy (RVH) and death. However, current therapies targeting pulmonary vascular remodeling and RVH remain poorly effective in reversing PAH. Overactivation of the protein tyrosine kinase Src plays an important role in tumor cell growth, proliferation and invasion; we thus hypothesized that inhibitors targeting Src activation could reverse experimental PAH. We demonstrated that Src was markedly activated in hypoxia-stimulated PASMCs from donors and PASMCs isolated from PAH patients. We investigated the effects of the Src-selective inhibitor 1-(1,1-dimethylethyl)-1-(4-methylphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine (PP1) and berberine (BBR) on PAH-PASMC proliferation and migration by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU) and wound-healing assays. Our in vitro results showed that inhibition of Src (Tyr416) phosphorylation repressed PAH-PASMC proliferation and migration by inhibiting hypoxia-inducible factor-1α (HIF-1α) expression through Akt/mTOR signal pathway. In vivo, PP1 and BBR significantly alleviated distal pulmonary vascular remodeling and decreased right ventricular systolic pressure (RVSP) and RVH in Sugen (SU) 5416/hypoxia (SU-PAH) mice. These findings demonstrate that pharmacological (PP1 or BBR) inhibition of Src activation could be a novel means of treating severe pulmonary vascular remodeling and RVH in PAH patients.


Asunto(s)
Hipertrofia Ventricular Derecha/tratamiento farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Familia-src Quinasas/genética , Animales , Berberina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/patología , Ratones , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Resistencia Vascular/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores
9.
JACC Cardiovasc Interv ; 12(3): 274-284, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30732732

RESUMEN

OBJECTIVES: The authors sought to assess the benefits of pulmonary artery denervation (PADN) among combined pre- and post-capillary pulmonary hypertension (CpcPH) patients in a prospective, randomized, sham-controlled trial. BACKGROUND: PADN has been shown to improve hemodynamics of pulmonary arterial hypertension in a series of patients. Additionally, benefits of targeted medical therapy for patients with CpcPH secondary to left-sided heart failure are unknown. METHODS: Ninety-eight CpcPH patients, defined as mean pulmonary arterial pressure ≥25 mm Hg, pulmonary capillary wedge pressure >15 mm Hg, and pulmonary vascular resistance (PVR) >3.0 Wood units (WU), were randomly assigned to PADN or sildenafil plus sham PADN. Standard medical therapy for heart failure was administered to all patients in both groups. The primary endpoint was the increase in the 6-min walk distance at the 6-month follow-up. The secondary endpoint was change in PVR. Clinical worsening was assessed in a post hoc analysis. The main safety endpoint was occurrence of pulmonary embolism. RESULTS: At 6 months, the mean increases in the 6-min walk distance were 83 m in the PADN group and 15 m in the sildenafil group (least square mean difference 66 m, 95% confidence interval: 38.2 to 98.8 m; p < 0.001). PADN treatment was associated with a significantly lower PVR than in the sildenafil group (4.2 ± 1.5 WU vs. 6.1 ± 2.9 WU; p = 0.001). Clinical worsening was less frequent in the PADN group compared with the sildenafil group (16.7% vs. 40%; p = 0.014). At the end of the study, there were 7 all-cause deaths and 2 cases of pulmonary embolism. CONCLUSIONS: PADN is associated with significant improvements in hemodynamic and clinical outcomes in patients with CpcPH. Further studies are warranted to define its precise role in the treatment of this patient population. (Pulmonary Arterial Denervation in Patients With Pulmonary Hypertension Associated With the Left Heart Failure [PADN-5]; NCT02220335).


Asunto(s)
Presión Arterial , Desnervación Autonómica , Tolerancia al Ejercicio , Insuficiencia Cardíaca/fisiopatología , Hipertensión Pulmonar/cirugía , Arteria Pulmonar/inervación , Prueba de Paso , Anciano , Desnervación Autonómica/efectos adversos , Desnervación Autonómica/mortalidad , China , Femenino , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/mortalidad , Humanos , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/mortalidad , Hipertensión Pulmonar/fisiopatología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Embolia Pulmonar/mortalidad , Embolia Pulmonar/fisiopatología , Recuperación de la Función , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
10.
J Cell Physiol ; 234(2): 1842-1850, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30132872

RESUMEN

BACKGROUND: Norepinephrine (NE)-mediated vasoconstriction plays an important role in pulmonary hypertension associated with left heart disease (PH-LHD). However, the role of NE-mediated endothelial cell dysfunction in the pathogenesis of PH-LHD remains to be elucidated. METHODS AND RESULTS: An enzyme-linked immunosorbent assay showed that the NE concentration in the plasma of patients with PH-LHD was higher and the nitrate-nitrite concentration was lower than those in the control group. NE treatment decreased phospho-Ser633-eNOS and ß2 -adrenergic receptor (ß2 -AR) levels in the membrane of human pulmonary artery endothelial cells (HPAECs) analysed by western blot analysis. Consistently, fluorescence microscopy and flow cytometry showed that nitric oxide (NO) production was also decreased in HPAECs. Coimmunoprecipitation confirmed a direct interaction between ß2 -AR and endothelial NO synthase (eNOS). Overexpression of ß2 -AR attenuated the decline in phospho-Ser633-eNOS and NO production. Additionally, the expression of phospho-Ser633-eNOS and ß2 -AR was decreased in human pulmonary artery endothelium. Finally, our results indicate that NE stimulated HPAEC proliferation, which was blocked by protein kinase A inhibitor or protein kinase B (PKB-AKT) inhibitor. CONCLUSIONS: These data provide a novel mechanism for NE-decreased endothelium-derived NO and NE-induced HPAEC proliferation that leads to PH-LHD, suggesting a potential therapeutic target for PH-LHD.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Hipertensión Pulmonar/metabolismo , Óxido Nítrico/metabolismo , Norepinefrina/farmacología , Arteria Pulmonar/efectos de los fármacos , Receptores Adrenérgicos beta 2/efectos de los fármacos , Disfunción Ventricular Izquierda/metabolismo , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Masculino , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Norepinefrina/sangre , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
11.
Pulm Circ ; 9(2): 2045894018816297, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30421645

RESUMEN

This study aimed to determine the benefits and correlated mechanisms of pulmonary artery denervation (PADN) for heart failure (HF) pulmonary hypertension (PH). PH secondary to HF is associated with poor clinical outcomes because there is no proper therapy for it. PADN showed improved outcomes for patients with HF-PH. However, the underlying mechanisms remain unknown. Supracoronary aortic banding (SAB) was used to create HF-PH models. Sprague-Dawley rats were randomly assigned to control, SAB, sham, SAB with PADN, and SAB without PADN groups. Surgical (longitudinally damaging vessel nerves) and chemical (10% phenol applied to the surface of nerves) PADN was performed for animals in the SAB with PADN group. Morphological, echocardiographic, hemodynamic, and protein expression changes were measured four weeks thereafter. Adrenergic receptor (AR) expressions of pulmonary arteries from four HF-PH patients and four patients without PH were measured. Ten HF-PH patients who underwent PADN were followed-up for six months. SAB-induced HF-PH was achieved by 50% of animals. Surgical and chemical PADN was associated with significant improvements in pulmonary artery muscularization, hemodynamics, and right ventricular functions. In pulmonary arterial specimens from HF-PH patients, ß2-AR and α1A/B-AR, as well as eNOS, were downregulated and α1D-AR was upregulated compared to those from patients without PH. PADN led to a mean increase of 84 m during the 6-min walk distance for HF-PH patients at six-month follow-up. HF-PH was characterized by downregulated ß2-AR, α1A-AR, and α1B-AR and by upregulated α1D-AR. PADN is associated with significant improvements in hemodynamics and pulmonary artery remodeling.

12.
J Cell Physiol ; 233(12): 9750-9762, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30078229

RESUMEN

Excessive proliferation, migration, and antiapoptosis of pulmonary artery (PA) smooth muscle cells (PASMCs) underlies the development of pulmonary vascular remodeling. The innervation of the PA is predominantly sympathetic, and increased levels of circulating catecholamines have been detected in pulmonary arterial hypertension (PAH), suggesting that neurotransmitters released by sympathetic overactivation may play an essential role in PAH. However, the responsible mechanism remains unclear. Here, to investigate the effects of norepinephrine (NE) on PASMCs and the related mechanism, we used 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, the proliferating cell nuclear antigen and the cell counting kit-8 assay to evaluate the proliferation of PASMCs, Boyden chamber migration, and wound-healing assays to assess migration and western blot analysis to investigate protein expression. We demonstrated that the phosphorylation level of the protein phosphatase 2A (PP2A) catalytic subunit (Y307) was higher in PAH patients and PAH models than in controls, both in vivo and in vitro. In addition, NE induced the proliferation and migration of PASMCs, which was attenuated by berberine (BBR), a Chinese herbal medicine, and/or PP2A overexpression. PP2A inhibition worsened NE-induced PAH and could not be reversed by BBR. Thus, PP2A is critical in driving PAH, and BBR may alleviate PAH via PP2A signaling pathways, thereby offering a potential therapeutic option for PAH.


Asunto(s)
Berberina/administración & dosificación , Hipertensión Pulmonar/tratamiento farmacológico , Proteína Fosfatasa 2/genética , Arteria Pulmonar/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Norepinefrina/toxicidad , Arteria Pulmonar/patología , Ratas , Transducción de Señal/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA