Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oxid Med Cell Longev ; 2022: 2555476, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35770049

RESUMEN

Pulmonary hypertension (PH) is caused by chronic hypoxia that induces the migration and proliferation of pulmonary arterial smooth muscle cells (PASMCs), eventually resulting in right heart failure. PH has been related to aberrant autophagy; however, the hidden mechanisms are still unclear. Approximately 40% East Asians, equivalent to 8% of the universal population, carry a mutation in Aldehyde dehydrogenase 2 (ALDH2), which leads to the aggregation of noxious reactive aldehydes and increases the propensity of several diseases. Therefore, we explored the potential aspect of ALDH2 in autophagy associated with PH. In vitro mechanistic studies were conducted in human PASMCs (HPASMCs) after lentiviral ALDH2 knockdown and treatment with platelet-derived growth factor-BB (PDGF-BB). PH was induced in wild-type (WT) and ALDH2-knockout (ALDH2-/-) mice using vascular endothelial growth factor receptor inhibitor SU5416 under hypoxic conditions (HySU). Right ventricular function was assessed using echocardiography and invasive hemodynamic monitoring. Histological and immunohistochemical analyses were performed to evaluate pulmonary vascular remodeling. EdU, transwell, and wound healing assays were used to evaluate HPASMC migration and proliferation, and electron microscopy and immunohistochemical and immunoblot assays were performed to assess autophagy. The findings demonstrated that ALDH2 deficiency exacerbated right ventricular pressure, hypertrophy, fibrosis, and right heart failure resulting from HySU-induced PH. ALDH2-/- mice exhibited increased pulmonary artery muscularization and 4-hydroxynonenal (4-HNE) levels in lung tissues. ALDH2 knockdown increased PDGF-BB-induced PASMC migration and proliferation and 4-HNE accumulation in vitro. Additionally, ALDH2 deficiency increased the number of autophagosomes and autophagic lysosomes together with autophagic flux and ERK1/2-Beclin-1 activity in lung tissues and PASMCs, indicating enhanced autophagy. In conclusion, the study shows that ALDH2 has a protective role against the migration and proliferation of PASMCs and PH, possibly by regulating autophagy through the ERK1/2-Beclin-1 pathway.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión Pulmonar , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa Mitocondrial/genética , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Animales , Autofagia , Becaplermina , Beclina-1/metabolismo , Proliferación Celular , Células Cultivadas , Insuficiencia Cardíaca/metabolismo , Hipertensión Pulmonar/genética , Sistema de Señalización de MAP Quinasas , Ratones , Miocitos del Músculo Liso/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Circulation ; 145(20): 1542-1556, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35430895

RESUMEN

BACKGROUND: Cardiac resident macrophages are self-maintaining and originate from embryonic hematopoiesis. After myocardial infarction, cardiac resident macrophages are responsible for the efficient clearance and degradation of apoptotic cardiomyocytes (efferocytosis). This process is required for inflammation resolution and tissue repair; however, the underlying molecular mechanisms remain unknown. Therefore, we aimed to identify the mechanisms of the continued clearance and degradation of phagolysosomal cargo by cardiac resident macrophages during myocardial infarction. METHODS: Multiple transgenic mice such as Lgmn-/-, LgmnF/F; LysMCre, LgmnF/F; Cx3cr1CreER, LgmnF/F; LyveCre, and cardiac macrophage Lgmn overexpression by adenovirus gene transfer were used to determine the functional significance of Lgmn in myocardial infarction. Immune cell filtration and inflammation were examined by flow cytometry and quantitative real-time polymerase chain reaction. Moreover, legumain (Lgmn) expression was analyzed by immunohistochemistry and quantitative real-time polymerase chain reaction in the cardiac tissues of patients with ischemic cardiomyopathy and healthy control subjects. RESULTS: We identified Lgmn as a gene specifically expressed by cardiac resident macrophages. Lgmn deficiency resulted in a considerable exacerbation in cardiac function, accompanied by the accumulation of apoptotic cardiomyocytes and a reduced index of in vivo efferocytosis in the border area. It also led to decreased cytosolic calcium attributable to defective intracellular calcium mobilization. Furthermore, the formation of LC3-II-dependent phagosome around secondary-encountered apoptotic cardiomyocytes was disabled. In addition, Lgmn deficiency increased infiltration of MHC-IIhigh CCR2+ macrophages and the enhanced recruitment of MHC-IIlow CCR2+ monocytes with downregulation of the anti-inflammatory mediators, interleukin-10, and transforming growth factor-ß and upregulationof the proinflammatory mediators interleukin-1ß, tumor necrosis factor-α, interleukin-6, and interferon-γ. CONCLUSIONS: Our results directly link efferocytosis to wound healing in the heart and identify Lgmn as a significant link between acute inflammation resolution and organ function.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Animales , Calcio/metabolismo , Cisteína Endopeptidasas , Humanos , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo
3.
Sci Adv ; 8(17): eabm3436, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35476440

RESUMEN

Macrophages play a vital role in cardiac repair following myocardial infarction (MI). An enriched environment (EE) is involved in the regulation of macrophage-related activities and disease progression; however, whether EE affects the phenotype and function of macrophages to improve postinfarction cardiac repair remains unknown. In this study, we found that EE improved cardiac function, decreased mortality, and ameliorated adverse ventricular remodeling in mice after MI, with these outcomes closely related to the increased survival of Ly6Clow macrophages and their CCR2-MHCIIlow subsets. EE increased the expression of brain-derived neurotrophic factor (BDNF) in the hypothalamus, leading to higher circulating levels of BDNF, which, in turn, regulated the cardiac macrophages. BDNF bound to tropomyosin receptor kinase B to activate downstream ERK1/2 and AKT pathways, promoting macrophage survival. These findings demonstrate that EE optimizes postinfarction cardiac repair and highlights the significance of EE as a previously unidentified strategy for impeding adverse ventricular remodeling.


Asunto(s)
Infarto del Miocardio , Remodelación Ventricular , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corazón , Macrófagos/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Miocardio/metabolismo
4.
Ann Transl Med ; 9(11): 926, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34350241

RESUMEN

BACKGROUND: To explore the source, the role and the specific mechanism of IL-35 and its downstream molecules in the development of pulmonary hypertension. METHODS: 8-10 weeks male mice were undergoing hypoxia combined with SU5416 (HySu) to establish a pulmonary hypertension (PH) model. The phenotype of PH mice was measured by immunohistochemistry and immunofluorescence staining. The levels of two subunits (EBI3 and p35 subunits) in lung tissue were measured by real-time PCR and western blotting. EBI3 monoclonal antibody was administrated as IL-35 neutralization to offset systemic IL-35 expression. Fludarabine, an inhibitor of STAT1 (signal transducer and activator of transcription 1) was used to clarify the role of STAT1 under IL-35 treatment. RESULTS: After pulmonary hypertension, the expression of IL-35 and its two subunits (EBI3 and p35 subunits) in lung tissue were significantly increased. And the two subunits of IL-35 are highly expressed in Treg cells. Compared with the controlled PH mice, the IL-35 neutralization PH mice showed aggravated pulmonary hypertension phenotype. The specific manifestations are the increase of right ventricular systolic pressure (RVSP), the growing proportion of right heart [RV/(LV+S)], and the remodeling of pulmonary blood vessels increases. The expression of pulmonary vascular endothelium (CD31) in PH mice increased, and the proliferation ability of vascular endothelium enhanced after IL-35 was inhibited. IL-35 phosphorylates STAT1 through the receptor GP130 on pulmonary vascular endothelial cells, which in turn inhibits endothelial cell proliferation. IL-35 recombinant protein can reduce the expression of CD31 in lung tissues of PH mice. But the administration of STAT1 inhibitor made it invalid from the IL-35 effect of reversing pulmonary hypertension. CONCLUSIONS: Tregs-derived IL-35 can reverse the remodeling of pulmonary blood vessels and alleviate the progression of pulmonary hypertension by reducing the proliferation of endothelial cells.

5.
Circulation ; 143(1): 45-61, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-32988222

RESUMEN

BACKGROUND: PCSK9 (proprotein convertase subtilisin/kexin 9), mainly secreted by the liver and released into the blood, elevates plasma low-density lipoprotein cholesterol by degrading low-density lipoprotein receptor. Pleiotropic effects of PCSK9 beyond lipid metabolism have been shown. However, the direct effects of PCSK9 on platelet activation and thrombosis, and the underlying mechanisms, as well, still remain unclear. METHODS: We detected the direct effects of PCSK9 on agonist-induced platelet aggregation, dense granule ATP release, integrin αIIbß3 activation, α-granule release, spreading, and clot retraction. These studies were complemented by in vivo analysis of FeCl3-injured mouse mesenteric arteriole thrombosis. We also investigated the underlying mechanisms. Using the myocardial infarction (MI) model, we explored the effects of PCSK9 on microvascular obstruction and infarct expansion post-MI. RESULTS: PCSK9 directly enhances agonist-induced platelet aggregation, dense granule ATP release, integrin αIIbß3 activation, P-selectin release from α-granules, spreading, and clot retraction. In line, PCSK9 enhances in vivo thrombosis in a FeCl3-injured mesenteric arteriole thrombosis mouse model, whereas PCSK9 inhibitor evolocumab ameliorates its enhancing effects. Mechanism studies revealed that PCSK9 binds to platelet CD36 and thus activates Src kinase and MAPK (mitogen-activated protein kinase)-extracellular signal-regulated kinase 5 and c-Jun N-terminal kinase, increases the generation of reactive oxygen species, and activates the p38MAPK/cytosolic phospholipase A2/cyclooxygenase-1/thromboxane A2 signaling pathways downstream of CD36 to enhance platelet activation, as well. Using CD36 knockout mice, we showed that the enhancing effects of PCSK9 on platelet activation are CD36 dependent. It is important to note that aspirin consistently abolishes the enhancing effects of PCSK9 on platelet activation and in vivo thrombosis. Last, we showed that PCSK9 activating platelet CD36 aggravates microvascular obstruction and promotes MI expansion post-MI. CONCLUSIONS: PCSK9 in plasma directly enhances platelet activation and in vivo thrombosis, and MI expansion post-MI, as well, by binding to platelet CD36 and thus activating the downstream signaling pathways. PCSK9 inhibitors or aspirin abolish the enhancing effects of PCSK9, supporting the use of aspirin in patients with high plasma PCSK9 levels in addition to PCSK9 inhibitors to prevent thrombotic complications.


Asunto(s)
Plaquetas/metabolismo , Antígenos CD36/metabolismo , Infarto del Miocardio/metabolismo , Activación Plaquetaria/fisiología , Proproteína Convertasa 9/metabolismo , Trombosis/metabolismo , Animales , Aspirina/farmacología , Aspirina/uso terapéutico , Plaquetas/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/tratamiento farmacológico , Inhibidores de PCSK9 , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/fisiología , Trombosis/tratamiento farmacológico
7.
Circ Res ; 127(10): 1323-1336, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32912104

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PAH) is characterized by progressive pulmonary vascular remodeling, accompanied by varying degrees of perivascular inflammation. Niacin, a commonly used lipid-lowering drug, possesses vasodilating and proresolution effects by promoting the release of prostaglandin D2 (PGD2). However, whether or not niacin confers protection against PAH pathogenesis is still unknown. OBJECTIVE: This study aimed to determine whether or not niacin attenuates the development of PAH and, if so, to elucidate the molecular mechanisms underlying its effects. METHODS AND RESULTS: Vascular endothelial growth factor receptor inhibitor SU5416 and hypoxic exposure were used to induce pulmonary hypertension (PH) in rodents. We found that niacin attenuated the development of this hypoxia/SU5416-induced PH in mice and suppressed progression of monocrotaline-induced and hypoxia/SU5416-induced PH in rats through the reduction of pulmonary artery remodeling. Niacin boosted PGD2 generation in lung tissue, mainly through H-PGDS (hematopoietic PGD2 synthases). Deletion of H-PGDS, but not lipocalin-type PGDS, exacerbated the hypoxia/SU5416-induced PH in mice and abolished the protective effects of niacin against PAH. Moreover, H-PGDS was expressed dominantly in infiltrated macrophages in lungs of PH mice and patients with idiopathic PAH. Macrophage-specific deletion of H-PGDS markedly decreased PGD2 generation in lungs, aggravated hypoxia/SU5416-induced PH in mice, and attenuated the therapeutic effect of niacin on PAH. CONCLUSIONS: Niacin treatment ameliorates the progression of PAH through the suppression of vascular remodeling by stimulating H-PGDS-derived PGD2 release from macrophages.


Asunto(s)
Antihipertensivos/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Hipolipemiantes/farmacología , Macrófagos/efectos de los fármacos , Niacina/farmacología , Animales , Antihipertensivos/uso terapéutico , Células Cultivadas , Humanos , Hipertensión Pulmonar/metabolismo , Hipolipemiantes/uso terapéutico , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macrófagos/metabolismo , Ratones , Niacina/uso terapéutico , Prostaglandina D2/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Ratas
8.
Cell Prolif ; 53(5): e12771, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32270546

RESUMEN

OBJECTIVE: Adipose-derived mesenchymal stem cells (ADSCs) offer great promise as cell therapy for ischaemic diseases. Due to their poor survival in the ischaemic environment, the therapeutic efficacy of ADSCs is still relatively low. Interleukin-11 (IL-11) has been shown to play a key role in promoting cell proliferation and protecting cells from oxidative stress injury. The aim of this study was to determine whether IL-11 could improve therapeutic efficacy of ADSCs in ischaemic diseases. METHODS AND RESULTS: ADSCs were prepared from inguinal subcutaneous adipose tissue and exposed to hypoxic environment. The protein expression of IL-11 was decreased after hypoxic treatment. In addition, ADSCs viability was increased after IL-11 treatment under hypoxia. Moreover, IL-11 enhanced ADSCs viability in a dose-dependent manner under normoxia. Importantly, IL-11 promoted ADSCs proliferation and migration and protected ADSCs against hydrogen peroxide-induced cellular death. Notably, IL-11 enhanced ADSCs proliferation and migration, also promoted cell survival and apoptosis resistance by STAT3 signalling. In vivo, mice were subjected to limb ischaemia and treated with IL-11 overexpression ADSCs and control ADSCs. IL-11 overexpression ADSCs improved perfusion recovery in the ischaemic muscles. CONCLUSIONS: We provide the evidence that IL-11 promoted ADSCs proliferation, stimulated ADSCs migration and attenuated ADSCs apoptosis by activation of STAT3 signalling. These results suggest that IL-11 facilitated ADSCs engraftment in ischaemic tissue, thereby enhanced ADSCs therapeutic efficacy.


Asunto(s)
Tejido Adiposo/metabolismo , Interleucina-11/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL
9.
Basic Res Cardiol ; 115(3): 28, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32236769

RESUMEN

This study aims to determine the effect of exercise on the cardiac function, metabolic profiles and related molecular mechanisms in mice with ischemic-induced heart failure (HF). HF was induced by myocardial infarction (MI) in C57BL6/N mice. Cardiac function and physical endurance were improved in HF mice after exercise. Micro-PET/CT scanning revealed enhanced myocardial glucose uptake in vivo in HF mice after exercise. Exercise reduced mitochondrial structural damage in HF mice. Cardiomyocytes isolated from HF + exercise mice showed increased glycolysis capacity, respiratory function and ATP production. Both mRNA and protein expression of glucose transporter 1 (GLUT1) were upregulated after exercise. Results of ChIP-PCR revealed a novel interaction between transcription factor myocyte enhancer factor 2a (MEF2a) and GLUT1 in hearts of HF + exercise mice. Exercise also activated myocardial AMP-activated protein kinase (AMPK), which in turn phosphorylated histone deacetylase 4 (HDAC4), and thereby modulated the GLUT1 expression through reducing its inhibition on MEF2a in HF mice. Inhibition of HDAC4 also improved cardiac function in HF mice. Moreover, knockdown of GLUT1 impaired the systolic and diastolic function of isolated cardiomyocytes. In conclusion, exercise improves cardiac function and glucose metabolism in HF mice through inhibiting HDAC4 and upregulating GLUT1 expression.


Asunto(s)
Transportador de Glucosa de Tipo 1/metabolismo , Histona Desacetilasas/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Condicionamiento Físico Animal/fisiología , Animales , Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Glucosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/rehabilitación , Regulación hacia Arriba
11.
Am J Respir Crit Care Med ; 201(10): 1263-1276, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31917615

RESUMEN

Rationale: Vascular remodeling, including smooth muscle cell hypertrophy and proliferation, is the key pathological feature of pulmonary arterial hypertension (PAH). Prostaglandin I2 analogs (beraprost, iloprost, and treprostinil) are effective in the treatment of PAH. Of note, the clinically favorable effects of treprostinil in severe PAH may be attributable to concomitant activation of DP1 (D prostanoid receptor subtype 1).Objectives: To study the role of DP1 in the progression of PAH and its underlying mechanism.Methods: DP1 levels were examined in pulmonary arteries of patients and animals with PAH. Multiple genetic and pharmacologic approaches were used to investigate DP1-mediated signaling in PAH.Measurements and Main Results: DP1 expression was downregulated in hypoxia-treated pulmonary artery smooth muscle cells and in pulmonary arteries from rodent PAH models and patients with idiopathic PAH. DP1 deletion exacerbated pulmonary artery remodeling in hypoxia-induced PAH, whereas pharmacological activation or forced expression of the DP1 receptor had the opposite effect in different rodent models. DP1 deficiency promoted pulmonary artery smooth muscle cell hypertrophy and proliferation in response to hypoxia via induction of mTORC1 (mammalian target of rapamycin complex 1) activity. Rapamycin, an inhibitor of mTORC1, alleviated the hypoxia-induced exacerbation of PAH in DP1-knockout mice. DP1 activation facilitated raptor dissociation from mTORC1 and suppressed mTORC1 activity through PKA (protein kinase A)-dependent phosphorylation of raptor at Ser791. Moreover, treprostinil treatment blocked the progression of hypoxia-induced PAH in mice in part by targeting the DP1 receptor.Conclusions: DP1 activation attenuates hypoxia-induced pulmonary artery remodeling and PAH through PKA-mediated dissociation of raptor from mTORC1. These results suggest that the DP1 receptor may serve as a therapeutic target for the management of PAH.


Asunto(s)
Hipoxia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Hipertensión Arterial Pulmonar/genética , Receptores Inmunológicos/genética , Receptores de Prostaglandina/genética , Remodelación Vascular/genética , Animales , Antihipertensivos/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Humanos , Hipertrofia , Inmunosupresores/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Hipertensión Arterial Pulmonar/metabolismo , Arteria Pulmonar , ARN Mensajero/metabolismo , Ratas , Sirolimus/farmacología
12.
J Mol Cell Cardiol ; 139: 1-13, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31958462

RESUMEN

OBJECTIVE: Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are related to in-stent-restenosis (ISR) following percutaneous coronary intervention (PCI). Osteoprotegerin (OPG) has been implicated in various vascular diseases. However, the effects of OPG on ISR and the underlying mechanism remained elusive. We here investigated the association between OPG and ISR, and to demonstrate the role and potential mechanisms of OPG in neointimal hyperplasia. APPROACH AND RESULTS: From 2962 patients who received coronary angiography and follow-up coronary angiography at approximately one year, 291 patients were diagnosed with ISR, and another 291 gender- and age- matched patients without ISR were selected as controls. Serum OPG levels were significantly increased in patients with ISR. Multivariable logistic regression analysis indicated that OPG level was independently associated with the increased risk of ISR. In a mouse femoral artery wire injury model, upregulated OPG was evidenced in vascular tissue after injury. OPG deletion attenuated the vascular injury-induced neointimal hyperplasia and related gene expression in mice. OPG promoted neointimal hyperplasia and human aortic smooth muscle cell (hASMC) proliferation and migration through activation of yes-associated protein (YAP), a major downstream effector of the Hippo signaling pathway, whereas knockdown or inhibition of YAP in hASMCs blunted OPG-induced above effects. Moreover, we found that OPG, as a ligand for integrin αVß3, mediated phosphorylation of focal adhesion kinase (FAK) and actin cytoskeleton reorganization, resulting in YAP dephosphorylation in hASMCs. OPG-dependent YAP and VSMC activation was prevented by treatment with αVß3-blocking antibodies and inhibitors of FAK and actin stress fibers. CONCLUSIONS: Increased serum OPG levels are associated with increased risk of ISR following PCI and OPG could promote neointimal hyperplasia in response to injury through integrin αVß3 mediated FAK and YAP activation, indicating OPG/YAP inhibition might serve as an attractive novel target for the prevention of ISR after PCI.


Asunto(s)
Reestenosis Coronaria/complicaciones , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Integrina alfaVbeta3/metabolismo , Neointima/complicaciones , Neointima/patología , Osteoprotegerina/metabolismo , Transducción de Señal , Stents/efectos adversos , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Anciano , Animales , Movimiento Celular , Proliferación Celular , Reestenosis Coronaria/sangre , Progresión de la Enfermedad , Femenino , Arteria Femoral/metabolismo , Arteria Femoral/patología , Humanos , Hiperplasia , Incidencia , Modelos Logísticos , Masculino , Ratones Endogámicos C57BL , Análisis Multivariante , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Neointima/sangre , Osteoprotegerina/sangre , Osteoprotegerina/deficiencia , Fosforilación/efectos de los fármacos , Índice de Severidad de la Enfermedad , Regulación hacia Arriba , Verteporfina/farmacología
13.
J Mol Cell Cardiol ; 137: 107-118, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31668970

RESUMEN

Cardiac fibrosis is a common feature of various cardiovascular diseases. Previous studies showed that acetaldehyde dehydrogenase 2 (ALDH2) deficiency exacerbated pressure overload-induced heart failure. However, the role and mechanisms of cardiac fibrosis in this process remain largely unknown. This study aimed to investigate the effect of ALDH2 deficiency on cardiac fibrosis in transverse aortic constriction (TAC) induced pressure overload model in mice. Echocardiography and histological analysis revealed cardiac dysfunction and enhanced cardiac fibrosis in TAC-operated animals; ALDH2 deficiency further aggravated these changes. ALDH2 chimeric mice were generated by bone marrow (BM) transplantation of WT mice into the lethally irradiated ALDH2KO mice. The proportion of circulating fibroblast progenitor cells (FPCs) and ROS level in BM after TAC were significantly higher in ALDH2KO mice than in ALDH2 chimeric mice. Furthermore, FPCs were isolated and cultured for in vitro mechanistic studies. The results showed that the stem cell-derived factor 1 (SDF-1)/C-X-C chemokine receptor 4 (CXCR4) axis played a major role in the recruitment of FPCs. In conclusion, our research reveals that increased bone marrow FPCs mobilization and myocardial homing contribute to the enhanced cardiac fibrosis and dysfunction induced by TAC in ALDH2 KO mice via exacerbating accumulation of ROS in BM and myocardial SDF-1 expression.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/deficiencia , Células de la Médula Ósea/patología , Fibroblastos/patología , Miocardio/patología , Células Madre/patología , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Animales , Animales Recién Nacidos , Polaridad Celular , Proliferación Celular , Quimiocina CXCL12/metabolismo , Constricción Patológica , Fibrosis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Estrés Oxidativo , Receptores CXCR4/metabolismo , Transducción de Señal
14.
Curr Pharm Des ; 25(35): 3751-3761, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31593529

RESUMEN

Exercise has long been recognized as a beneficial living style for cardiovascular health. It has been applied to be a central component of cardiac rehabilitation for patients with chronic heart failure (CHF), coronary heart disease (CHD), post-acute coronary syndrome (ACS) or primary percutaneous coronary intervention (PCI), post cardiac surgery or transplantation. Although the effect of exercise is multifactorial, in this review, we focus on the specific contribution of regular exercise on the heart and vascular system. We will summarize the known result of clinical findings and possible mechanisms of chronic exercise on the cardiovascular system.


Asunto(s)
Síndrome Coronario Agudo/terapia , Rehabilitación Cardiaca/métodos , Enfermedad Coronaria/terapia , Terapia por Ejercicio , Insuficiencia Cardíaca/terapia , Procedimientos Quirúrgicos Cardíacos , Trasplante de Corazón , Humanos , Intervención Coronaria Percutánea
15.
J Mol Cell Cardiol ; 134: 119-130, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31299216

RESUMEN

BACKGROUND: Trimethylamine N-oxide (TMAO), a gut microbe-derived metabolite of dietary choline and other trimethylamine-containing nutrients, has been associated with poor prognosis in coronary heart disease. However, the role and underlying mechanisms of TMAO in the cardiac fibrosis after myocardial infarction (MI) remains unclear. METHODS: We used mouse MI models and primary cardiac fibroblasts cultures to study the role of TMAO in the heart and in cardiac fibroblasts. C57BL/6 mice were fed a control diet, high choline (1.2%) or/and DMB diet or a diet containing TMAO (0.12%) starting 3 weeks before MI. DMB, a structural analogue of choline, inhibited microbial TMA lyases and reduced the level of TMAO in mice. Cardiac function was measured 7 days after MI using echocardiography. One week post MI, myocardial tissues were collected to evaluate cardiac fibrosis, and blood samples were evaluated for TMAO levels. The expression of TGF-ß receptor, P-Smad2, α-SMA or collagen I in myocardial tissues and fibroblasts were analyzed by western blot or immunocytochemistry. RESULTS: We demonstrated that cardiac function and cardiac fibrosis were significantly deteriorated in mice fed either TMAO or high choline diets compared with the control diet, and DMB reversed the cardiac function damage of high choline diet (p < .05). Cardiomyocyte necrosis, apoptosis and macrophage infiltration after MI was significantly increased after treatment with TMAO or high choline diets. The size and migration of fibroblasts were increased after TMAO treatment compared with non-treated fibroblasts in vitro. Furthermore, TMAO increased TGF-ß receptor I expression, which promoted the phosphorylation of Smad2 and up-regulated the expression of α-SMA and collagen I. The ubiquitination of TGF-ßRI was decreased in neonatal mouse fibroblasts after TMAO treatment. TMAO also inhibited the expression of smurf2. Inhibition of TGF-ß1 receptor with the small molecule inhibitor SB431542 decreased TGF-ß receptor I expression, reduced the phosphorylation of Smad2, down-regulated TMAO-induced α-SMA and collagen I expression in cardiac fibroblasts. CONCLUSIONS: Cardiac function and cardiac fibrosis were significantly exacerbated in mice fed diets supplemented with either choline or TMAO, probably through accelerating the transformation of fibroblasts into myofibroblasts, indicating activation of TGF-ßRI/Smad2 pathway.


Asunto(s)
Diferenciación Celular/fisiología , Fibroblastos/metabolismo , Fibrosis/metabolismo , Microbioma Gastrointestinal/fisiología , Metilaminas/metabolismo , Miocardio/metabolismo , Miofibroblastos/metabolismo , Animales , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
17.
Circ Res ; 124(9): 1323-1336, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30832557

RESUMEN

RATIONALE: Targeting inflammation has been shown to provide clinical benefit in the field of cardiovascular diseases. Although manipulating regulatory T-cell function is an important goal of immunotherapy, the molecules that mediate their suppressive activity remain largely unknown. IL (interleukin)-35, an immunosuppressive cytokine mainly produced by regulatory T cells, is a novel member of the IL-12 family and is composed of an EBI3 (Epstein-Barr virus-induced gene 3) subunit and a p35 subunit. However, the role of IL-35 in infarct healing remains elusive. OBJECTIVE: This study aimed to determine whether IL-35 signaling is involved in healing and cardiac remodeling after myocardial infarction (MI) and, if so, to elucidate the underlying molecular mechanisms. METHODS AND RESULTS: IL-35 subunits (EBI3 and p35), which are mainly expressed in regulatory T cells, were upregulated in mice after MI. After IL-35 inhibition, mice showed impaired infarct healing and aggravated cardiac remodeling, as demonstrated by a significant increase in mortality because of cardiac rupture, decreased wall thickness, and worse cardiac function compared with wild-type MI mice. IL-35 inhibition also led to decreased expression of α-SMA (α-smooth muscle actin) and collagen I/III in the hearts of mice after MI. Pharmacological inhibition of IL-35 suppressed the accumulation of Ly6Clow and major histocompatibility complex IIlow/C-C motif chemokine receptor type 2- (MHC IIlow CCR2-) macrophages in infarcted hearts. IL-35 activated transcription of CX3CR1 (C-X3-C motif chemokine receptor 1) and TGF (transforming growth factor) ß1 in macrophages by inducing GP130 signaling, via IL12Rß2 and phosphorylation of STAT1 (signal transducer and activator of transcription family) and STAT4 and subsequently promoted Ly6Clow macrophage survival and extracellular matrix deposition. Moreover, compared with control MI mice, IL-35-treated MI mice showed increased expression of α-SMA and collagen within scars, correlating with decreased left ventricular rupture rates. CONCLUSIONS: IL-35 reduces cardiac rupture, improves wound healing, and attenuates cardiac remodeling after MI by promoting reparative CX3CR1+Ly6Clow macrophage survival.


Asunto(s)
Interleucinas/fisiología , Macrófagos/fisiología , Infarto del Miocardio/fisiopatología , Cicatrización de Heridas/fisiología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/farmacología , Receptor 1 de Quimiocinas CX3C/biosíntesis , Receptor 1 de Quimiocinas CX3C/genética , Supervivencia Celular , Cicatriz/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Regulación de la Expresión Génica/fisiología , Rotura Cardíaca Posinfarto/fisiopatología , Rotura Cardíaca Posinfarto/prevención & control , Interleucinas/antagonistas & inhibidores , Interleucinas/biosíntesis , Interleucinas/genética , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor/biosíntesis , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Miocardio/metabolismo , Receptores de Citocinas/antagonistas & inhibidores , Receptores de Citocinas/biosíntesis , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Regulación hacia Arriba , Remodelación Ventricular/fisiología
18.
Arterioscler Thromb Vasc Biol ; 39(4): e130-e145, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30676070

RESUMEN

Objective- Macrophages participate in the pathogenesis of pulmonary arterial hypertension (PAH). Lgmn (Legumain), a newly discovered cysteine proteinase belonging to the C13 peptidase family, is primarily expressed in macrophages; however, its roles in PAH remain unknown. Approach and Results- Herein, Lgmn was upregulated in lung tissues of PAH mice subjected to hypoxia plus SU5416 and PAH rats challenged with monocrotaline. Global Lgmn ablation and macrophage-specific ablation alleviated PAH compared with wild-type mice, evident from a reduction in right ventricular systolic pressure, the ratio of the right ventricular wall to the left ventricular wall plus the septum, the pulmonary vascular media thickness, and pulmonary vascular muscularization. Increased expression of ECM (extracellular matrix) proteins was correlated with MMP (matrix metalloproteinase)-2 activation and TGF (transforming growth factor)-ß1 signaling in the PAs. Although Lgmn did not affect inflammatory cell infiltration and PA smooth muscle cell proliferation, it drove increased the synthesis of ECM proteins via MMP-2 activation. MMP-2 hydrolyzed the TGF-ß1 precursor to the active form. An Lgmn-specific inhibitor markedly ameliorated PAH. Clinically, serum Lgmn levels were closely associated with the severity of idiopathic PAH. Conclusions- Our results indicate that Lgmn inhibition could be an effective strategy for preventing or delaying PAH.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Hipertensión Pulmonar/enzimología , Macrófagos/enzimología , Metaloproteinasa 2 de la Matriz/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Animales , Inhibidores de Caspasas/farmacología , Cisteína Endopeptidasas/deficiencia , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/prevención & control , Hipoxia/enzimología , Indoles/toxicidad , Inflamación , Pulmón/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Monocrotalina/toxicidad , Pirroles/toxicidad , Ratas , Índice de Severidad de la Enfermedad , Transducción de Señal , Remodelación Vascular/fisiología
19.
J Exp Med ; 215(8): 2175-2195, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29970474

RESUMEN

Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by progressive pulmonary artery (PA) remodeling. T helper 2 cell (Th2) immune response is involved in PA remodeling during PAH progression. Here, we found that CRTH2 (chemoattractant receptor homologous molecule expressed on Th2 cell) expression was up-regulated in circulating CD3+CD4+ T cells in patients with idiopathic PAH and in rodent PAH models. CRTH2 disruption dramatically ameliorated PA remodeling and pulmonary hypertension in different PAH mouse models. CRTH2 deficiency suppressed Th2 activation, including IL-4 and IL-13 secretion. Both CRTH2+/+ bone marrow reconstitution and CRTH2+/+ CD4+ T cell adoptive transfer deteriorated hypoxia + ovalbumin-induced PAH in CRTH2-/- mice, which was reversed by dual neutralization of IL-4 and IL-13. CRTH2 inhibition alleviated established PAH in mice by repressing Th2 activity. In culture, CRTH2 activation in Th2 cells promoted pulmonary arterial smooth muscle cell proliferation through activation of STAT6. These results demonstrate the critical role of CRTH2-mediated Th2 response in PAH pathogenesis and highlight the CRTH2 receptor as a potential therapeutic target for PAH.


Asunto(s)
Hipertensión Pulmonar/inmunología , Activación de Linfocitos/inmunología , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Células Th2/inmunología , Traslado Adoptivo , Adulto , Animales , Anticuerpos/farmacología , Presión Sanguínea/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Quimera , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Humanos , Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Inmunidad/efectos de los fármacos , Indoles , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Ovalbúmina , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Pirroles , Receptores Inmunológicos/deficiencia , Receptores de Prostaglandina/deficiencia , Factor de Transcripción STAT6/metabolismo , Regulación hacia Arriba/efectos de los fármacos
20.
Cardiovasc Res ; 113(6): 586-597, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28407046

RESUMEN

AIMS: Extracellular matrix (ECM) proteins accumulation contributes to the progression of pulmonary arterial hypertension (PAH), a rare and fatal cardiovascular condition defined by high pulmonary arterial pressure, whether primary, idiopathic, or secondary to other causes. The receptor for advanced glycation end products (RAGE) is constitutively expressed in the lungs and plays an important role in ECM deposition. Nonetheless, the mechanisms by which RAGE mediates ECM deposition/formation in pulmonary arteries and its roles in PAH progression remain unclear. METHODS AND RESULTS: Expression of RAGE and its activating ligands, S100/calgranulins and high mobility group box 1 (HMGB1), were increased in both human and mouse pulmonary arterial smooth muscle cells (PASMCs) under hypoxic conditions and were also strikingly upregulated in pulmonary arteries in hypoxia plus SU5416 (HySu)-induced PAH in mice. RAGE deletion alleviated pulmonary arterial pressure and restrained extracellular matrix accumulation in pulmonary arteries in HySu-induced PAH murine model. Moreover, blocking RAGE activity with a neutralizing antibody in human PASMCs, or RAGE deficiency in mouse PASMCs exposed to hypoxia, suppressed the expression of fibrotic proteins by reducing TGF-ß1 expression. RAGE reconstitution in deficient mouse PASMCs restored hypoxia-stimulated TGF-ß1 production via ERK1/2 and p38 MAPK pathway activation and subsequently increased ECM protein expression. Interestingly, HMGB1 acting on RAGE, not toll-like receptor 4 (TLR4), induced ECM deposition in PASMCs. Finally, in both idiopathic PAH patients and HySu-induced PAH mice, soluble RAGE (sRAGE) levels in serum were significantly elevated compared to those in controls. CONCLUSIONS: Activation of RAGE facilitates the development of hypoxia-induced pulmonary hypertension by increase of ECM deposition in pulmonary arteries. Our results indicate that sRAGE may be a potential biomarker for PAH diagnosis and disease severity, and that RAGE may be a promising target for PAH treatment.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Hipertensión Pulmonar/inducido químicamente , Indoles , Arteria Pulmonar/metabolismo , Pirroles , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Remodelación Vascular , Animales , Estudios de Casos y Controles , Hipoxia de la Célula , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Ligandos , Masculino , Persona de Mediana Edad , Fosforilación , Arteria Pulmonar/patología , Interferencia de ARN , Receptor para Productos Finales de Glicación Avanzada/genética , Subunidad beta de la Proteína de Unión al Calcio S100/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...