Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.334
Filtrar
1.
Oxid Med Cell Longev ; 2022: 6603296, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35096272

RESUMEN

CD147, also known as EMMPRIN or basigin, is a transmembrane glycoprotein receptor that activates matrix metalloproteinases and promotes inflammation. CD147 function is regulated by posttranslational modifications of which glycosylation has attracted the most attention. In this study, we demonstrated that glycosylated CD147 was the dominant form in heart tissue, and its levels were markedly elevated in response to transverse aortic constriction (TAC). Adeno-associated virus 9-mediated, cardiac-specific overexpression of wild-type CD147 in mice significantly promoted pressure overload-induced pathological cardiac remodeling accompanied by augmented oxidative stress and ferroptosis. By contrast, mutations of CD147 glycosylation sites notably weakened these detrimental effects of CD147. Mechanistically, CD147 exacerbated TAC-induced pathological cardiac remodeling via direct binding with the adaptor molecule TRAF2 and subsequent activation of TAK1 signalling, which was dependent on glycosylation of CD147. Collectively, our findings provide the first evidence that CD147 promoted pathological cardiac remodeling and dysfunction in a glycosylation-dependent manner through binding the adaptor protein TRAF2 and activating the downstream TRAF2-TAK1 signalling pathway. Thus, glycosylation of CD147 may be a potent interventional target for heart failure treatment.


Asunto(s)
Basigina/efectos adversos , Cardiomegalia/fisiopatología , Animales , Glicosilación , Humanos , Masculino , Ratones
2.
Cell Tissue Res ; 387(1): 111-121, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34669021

RESUMEN

Cardiac hypertrophy is considered as a common pathophysiological process in various cardiovascular diseases. CUG triplet repeat-binding protein 1 (CELF1) is an RNA-binding protein that has been shown to be an important post-transcription regulator and involved in several types of cancer, whereas its role in cardiac remodeling remains unclear. Herein, we found that the expression of CELF1 was significantly increased in pressure overload-induced hypertrophic hearts and angiotensin II (Ang II)-induced neonatal cardiomyocytes. Based on transverse aortic constriction-induced cardiac hypertrophy model, CELF1 deficiency markedly ameliorated cardiac hypertrophy, cardiac fibrosis, oxidative stress, and apoptosis. Accordingly, CELF1 deficiency alleviated the production of reactive oxygen species (ROS) and apoptosis of neonatal cardiomyocytes via inhibition of Raf1, TAK1, ERK1/2, and p38 phosphorylation. Mechanistically, depletion or overexpression of CELF1 negatively regulated the protein expression of phosphatidylethanolamine-binding protein 1 (PEBP1), while the mRNA expression of PEBP1 remained unchanged. RNA immunoprecipitation revealed that CELF1 directly interacted with PEBP1 mRNA. Biotin pull-down analysis and dual-luciferase assay showed that CELF1 directly bound to the fragment 1 within 3'UTR of PEBP1. Moreover, knockdown of PEBP1 partially enhanced the production of ROS and apoptosis of neonatal cardiomyocytes inhibited by CELF1 deficiency. In conclusion, CELF1 binds to the 3'UTR of PEBP1 and acts as an endogenous activator of MAPK signaling pathway. Inhibition of CELF1 attenuates pathological cardiac hypertrophy, oxidative stress, and apoptosis, thus could be a potential therapeutic strategy of pathological cardiac hypertrophy.


Asunto(s)
Proteínas CELF1/metabolismo , Cardiomegalia/genética , Ecocardiografía/métodos , Miocitos Cardíacos/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Cardiomegalia/fisiopatología , Humanos , Ratones , Transducción de Señal
3.
Int J Mol Sci ; 22(21)2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34769252

RESUMEN

Involvement of the Toll-like receptor 4 (TLR4) in maladaptive cardiac remodeling and heart failure (HF) upon pressure overload has been studied extensively, but less is known about the role of TLR2. Interplay and redundancy of TLR4 with TLR2 have been reported in other organs but were not investigated during cardiac dysfunction. We explored whether TLR2 deficiency leads to less adverse cardiac remodeling upon chronic pressure overload and whether TLR2 and TLR4 additively contribute to this. We subjected 35 male C57BL/6J mice (wildtype (WT) or TLR2 knockout (KO)) to sham or transverse aortic constriction (TAC) surgery. After 12 weeks, echocardiography and electrocardiography were performed, and hearts were extracted for molecular and histological analysis. TLR2 deficiency (n = 14) was confirmed in all KO mice by PCR and resulted in less hypertrophy (heart weight to tibia length ratio (HW/TL), smaller cross-sectional cardiomyocyte area and decreased brain natriuretic peptide (BNP) mRNA expression, p < 0.05), increased contractility (QRS and QTc, p < 0.05), and less inflammation (e.g., interleukins 6 and 1ß, p < 0.05) after TAC compared to WT animals (n = 11). Even though TLR2 KO TAC animals presented with lower levels of ventricular TLR4 mRNA than WT TAC animals (13.2 ± 0.8 vs. 16.6 ± 0.7 mg/mm, p < 0.01), TLR4 mRNA expression was increased in animals with the largest ventricular mass, highest hypertrophy, and lowest ejection fraction, leading to two distinct groups of TLR2 KO TAC animals with variations in cardiac remodeling. This variation, however, was not seen in WT TAC animals even though heart weight/tibia length correlated with expression of TLR4 in these animals (r = 0.078, p = 0.005). Our data suggest that TLR2 deficiency ameliorates adverse cardiac remodeling and that ventricular TLR2 and TLR4 additively contribute to adverse cardiac remodeling during chronic pressure overload. Therefore, both TLRs may be therapeutic targets to prevent or interfere in the underlying molecular processes.


Asunto(s)
Presión Sanguínea , Cardiomegalia/metabolismo , Ventrículos Cardíacos/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Remodelación Ventricular , Animales , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Masculino , Ratones , Ratones Noqueados , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
4.
Cells ; 10(10)2021 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-34685620

RESUMEN

In the present study, we investigated a novel signaling target in diabetic cardiomyopathy where inflammation induces caspase-1-dependent cell death, pyroptosis, involving Nek7-GBP5 activators to activate the NLRP3 inflammasome, destabilizes cardiac structure and neovascularization. Furthermore, we explored the therapeutic ability of bone morphogenetic protein-7 (BMP-7) to attenuate these adverse effects. C57BL/6J mice (n = 16 mice/group) were divided into: control (200 mg/kg, 0.9% saline intraperitoneal injection, i.p.); Streptozotocin (STZ) and STZ-BMP-7 groups (STZ, 200 mg/kg, i.p. injection). After 6 weeks, heart function was examined with echocardiography, and mice were sacrificed. Immunostaining, Western blotting, H&E, and Masson's trichrome staining was performed on heart tissues. STZ-induced diabetic cardiomyopathy significantly increased inflammasome formation (TLR4, NLRP3, Nek7, and GBP5), pyroptosis markers (caspase-1, IL-1ß, and IL-18), inflammatory cytokines (IL-6 and TNF-α), MMP9, and infiltration of monocytes (CD14), macrophage (iNOS), and dendritic cells (CD11b and CD11c) (p < 0.05). Moreover, a significant endothelial progenitor cells (EPCs) dysfunction (c-Kit/FLk-1, CD31), adverse cardiac remodeling, and reduction in left ventricular (LV) heart function were observed in STZ versus control (p < 0.05). Treatment with BMP-7 significantly reduced inflammasome formation, pyroptosis, and inflammatory cytokines and infiltrated inflammatory cells. In addition, BMP-7 treatment enhanced EPC markers and neovascularization and subsequently improved cardiac remodeling in a diabetic heart. Moreover, a significant improvement in LV heart function was achieved after BMP-7 administration relative to diabetic mice (p < 0.05). In conclusion, BMP-7 attenuated inflammation-induced pyroptosis, adverse cardiac remodeling, and improved heart function via the TLR4-NLRP3 inflammasome complex activated by novel signaling Nek7/GBP5. Our BMP-7 pre-clinical studies of mice could have significant potential as a future therapy for diabetic patients.


Asunto(s)
Proteína Morfogenética Ósea 7/farmacología , Cardiomiopatías Diabéticas/patología , Inflamación/patología , Miocardio/patología , Piroptosis , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 7/uso terapéutico , Cardiomegalia/complicaciones , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Caspasa 1/metabolismo , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Cardiomiopatías Diabéticas/complicaciones , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Células Endoteliales/metabolismo , Fibrosis , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neovascularización Fisiológica , Tamaño de los Órganos/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Piroptosis/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Función Ventricular Izquierda
5.
Biomed Pharmacother ; 143: 112194, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34563949

RESUMEN

Cardiac pressure overload is a crucial risk factor for cardiac hypertrophy and heart failure. Our previous study showed that depletion of the ß3-adrenergic receptor (ADRB3) induced left ventricular diastolic dysfunction via potential regulation of energy metabolism and cardiac contraction. However, the effects of ADRB3 on pressure overload-induced heart failure remain unclear. In the present study, systemic ADRB3-knockout mice suffering from transverse aortic constriction (TAC) surgery were used to identify the effects of ADRB3 on pressure overload-induced heart failure. Compared to wild-type mice, ADRB3 depletion significantly improved the left ventricular ejection fraction, reduced left ventricular posterior wall thickness and interventricular septum thickness, and decreased the area of cardiomyocytes after TAC. RNA sequencing and bioinformatics analysis showed that ADRB3 depletion up-regulated 275 mRNAs and down-regulated 105 mRNAs in mice suffering TAC surgery. GO analysis, GO-tree analysis, and GSEA showed that ADRB3 depletion mainly enhanced the innate immune response of hearts in cardiac pressure overload mice. In addition, pathway analysis and Pathway-Act analysis presented that innate immune response-related pathways, including RIG-I-like receptor signaling pathway, antigen processing and presentation, Toll-like receptor signaling pathway, and cell adhesion molecules, were significantly enriched in ADRB3-KO-TAC mice. Ten hub genes were identified using protein-protein interaction network, MCODE, and cytoHubba analysis. Furthermore, the depletion and activation of ADRB3 validated the effects of ADRB3 on the innate immune response of hearts after TAC. In conclusion, ADRB3 depletion relieves pressure overload-induced cardiac hypertrophy and heart failure, and these effects could be explained by the enhancement of innate immune response.


Asunto(s)
Cardiomegalia/prevención & control , Insuficiencia Cardíaca/prevención & control , Inmunidad Innata , Miocardio/metabolismo , Receptores Adrenérgicos beta 3/deficiencia , Animales , Cardiomegalia/inmunología , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Inmunidad Innata/genética , Masculino , Ratones Noqueados , Miocardio/inmunología , Mapas de Interacción de Proteínas , Receptores Adrenérgicos beta 3/genética , Transducción de Señal , Volumen Sistólico , Transcriptoma , Función Ventricular Izquierda , Remodelación Ventricular
6.
Aging (Albany NY) ; 13(18): 22516-22527, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34582362

RESUMEN

Cardiac fibrosis could induce abnormal cardiac function and become a novel target for cardiac hypertrophy and chronic heart failure. MiRNA-320 is a crucial miRNA in cardiovascular disease, but it is poorly understood whether it plays a role in cardiac fibrosis pathogenesis. We aimed to identify the specific underlying mechanism of miR-320 in cardiac fibrosis and hypertrophic pathogenesis. In our study, the GEO datasets revealed that STAT3 was significantly highly expressed in cardiomyocyte lines. MiR-320 activation and STAT3 signaling pathways were statistically significantly connected. Furthermore, miR-320 was highly associated with cardiac fibrosis and hypertrophic disease. Interstitial fibrosis was observed in the mice subjected to TAC surgery, markedly enhanced in miR-320 mimics. Mechanistically, we revealed that miR-320 mimics aggravated the pressure overload and induced cardiac hypertrophy and fibrosis via the IL6/STAT3/PTEN axis. MiR-320 mimics accelerated cardiac hypertrophy and cardiac fibrosis via the IL6/STAT3/PTEN axis. These results suggest that targeting miR-320 may represent a potential therapeutic strategy for cardiac hypertrophy and fibrosis.


Asunto(s)
Cardiomegalia/fisiopatología , Insuficiencia Cardíaca/metabolismo , Interleucina-6/metabolismo , Miocitos Cardíacos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Células Cultivadas , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Fibrosis , Ratones , MicroARNs
7.
Bioengineered ; 12(1): 5491-5503, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34506248

RESUMEN

Excessive oxidative stress, inflammation, and myocardial hypertrophy have been associated with diabetic cardiomyopathy (DCM). S14G-humanin (HNG) is a potent humanin analogue that has demonstrated cytoprotective effects in a variety of cells and tissues. However, the pharmacological function of HNG in diabetic cardiomyopathy has not yet been reported. In the present study, we investigated the protective effects of HNG against streptozotocin (STZ)-induced cardiac dysfunction in diabetic mice. Myocardial hypertrophy in diabetic mice was determined using Wheat Gem Agglutinin (WGA) staining. The heart function was measured with Echocardiographic imaging. Levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) proteins in plasma were measured using enzyme-linked immunosorbent assay (ELISA) kits. Protein expression of Phosphorylated p38/p38 was determined using western blots. We found that HNG treatment attenuated the STZ-induced myocardial hypertrophy and significantly improved heart function. Also, its treatment proved effective as it reduced the levels of several myocardial injury indicators, including creatine kinase-MB (CK-MB), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and both the cardiac and plasma levels of TNF-α and IL-6, highlighting its effect on the STZ-induced myocardial injury. Lastly, HNG suppressed the activation of the p38/nuclear factor kappa-B (NF-κB) signaling pathway. S14G humanin possesses protective effects against streptozotocin-induced cardiac dysfunction through inhibiting the activation of the p38/NF-κB signaling pathway.


Asunto(s)
Cardiomegalia , Corazón/efectos de los fármacos , Péptidos/farmacología , Sustancias Protectoras/farmacología , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Diabetes Mellitus Experimental/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estreptozocina/efectos adversos
8.
Sci Rep ; 11(1): 18102, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34518583

RESUMEN

Arteriovenous fistula (AVF) is the preferred type of vascular access for maintenance haemodialysis but it may contribute to maladaptive cardiovascular remodelling. We studied the effect of AVF creation on cardiac structure and function in patients with chronic kidney disease (CKD). In this prospective cohort study patients with CKD listed for first AVF creation underwent cardiac magnetic resonance (CMR) imaging at baseline and at 6 weeks. All participants had ultrasound measurements of fistula blood flow at 6 weeks. The primary outcome was the change in left ventricular (LV) mass. Secondary outcomes included changes in LV volumes, LV ejection fraction, cardiac output, LV global longitudinal strain and N-terminal-pro B-type natriuretic peptide (NT-proBNP). A total of 55 participants were enrolled, of whom 40 (mean age 59 years) had AVF creation and completed both scans. On the second CMR scan, a mean increase of 7.4 g (95% CI 1.1-13.7, p = 0.02) was observed in LV mass. Significant increases in LV end-diastolic volumes (p = 0.04) and cardiac output (p = 0.02) were also seen after AVF creation. No significant changes were observed in LV end-systolic volumes, LV ejection fraction, NT-proBNP and LV global longitudinal strain. In participants with fistula blood flows ≥ 600 mL/min (n = 22) the mean increase in LV mass was 15.5 g (95% CI 7.3-23.8) compared with a small decrease of 2.5 g (95% CI - 10.6 to 5.6) in participants with blood flows < 600 mL/min (n = 18). Creation of AVF for haemodialysis resulted in a significant increase of LV myocardial mass within weeks after surgery, which was proportional to the fistula flow.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Cardiopatías/diagnóstico , Cardiopatías/etiología , Hemodinámica , Diálisis Renal/efectos adversos , Anciano , Derivación Arteriovenosa Quirúrgica/métodos , Gasto Cardíaco , Cardiomegalia/diagnóstico , Cardiomegalia/etiología , Cardiomegalia/fisiopatología , Manejo de la Enfermedad , Ecocardiografía , Femenino , Cardiopatías/terapia , Pruebas de Función Cardíaca , Humanos , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/terapia , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Pronóstico , Diálisis Renal/métodos , Resultado del Tratamiento
9.
PLoS One ; 16(8): e0255022, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34339435

RESUMEN

Prolyl hydroxylase (PH) enzymes control the degradation of hypoxia-inducible factor (HIF), a transcription factor known to regulate erythropoiesis, angiogenesis, glucose metabolism, cell proliferation, and apoptosis. HIF-PH inhibitors (HIF-PHIs) correct anemia in patients with renal disease and in animal models of anemia and kidney disease. However, the effects of HIF-PHIs on comorbidities associated with kidney disease remain largely unknown. We evaluated the effects of the HIF-PHI FG-2216 in obese ZSF1 (Ob-ZSF1) rats, an established model of kidney failure with metabolic syndrome. Following unilateral nephrectomy (Nx) at 8 weeks of age, rats were treated with 40 mg/kg FG-2216 or vehicle by oral gavage three times per week for up to 18 weeks. FG-2216 corrected blood hemoglobin levels and improved kidney function and histopathology in Nx-Ob-ZSF1 rats by increasing the glomerular filtration rate, decreasing proteinuria, and reducing peritubular fibrosis, tubular damage, glomerulosclerosis and mesangial expansion. FG-2216 increased renal glucose excretion and decreased body weight, fat pad weight, and serum cholesterol in Nx-Ob-ZSF1 rats. Additionally, FG-2216 corrected hypertension, improved diastolic and systolic heart function, and reduced cardiac hypertrophy and fibrosis. In conclusion, the HIF-PHI FG-2216 improved renal and cardiovascular outcomes, and reduced obesity in a rat model of kidney disease with metabolic syndrome. Thus, in addition to correcting anemia, HIF-PHIs may provide renal and cardiac protection to patients suffering from kidney disease with metabolic syndrome.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Inhibidores de Prolil-Hidroxilasa/uso terapéutico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Animales , Biomarcadores/sangre , Cardiomegalia/sangre , Cardiomegalia/complicaciones , Cardiomegalia/fisiopatología , Cardiomiopatías/sangre , Cardiomiopatías/complicaciones , Cardiomiopatías/fisiopatología , Glucosa/metabolismo , Hemoglobinas/metabolismo , Hipertensión/sangre , Hipertensión/complicaciones , Hipertensión/fisiopatología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Riñón/fisiopatología , Enfermedades Renales/sangre , Enfermedades Renales/complicaciones , Enfermedades Renales/fisiopatología , Masculino , Obesidad/sangre , Obesidad/complicaciones , Obesidad/fisiopatología , Inhibidores de Prolil-Hidroxilasa/farmacología , Ratas , Bibliotecas de Moléculas Pequeñas/farmacología
10.
Cell Biol Int ; 45(11): 2357-2367, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34370360

RESUMEN

Cardiac hypertrophy has been a high prevalence rate throughout the world. It has posed a big threat to public health due to limited therapeutic approaches. Previous studies showed that pathological cardiac hypertrophy was associated with autophagy, microRNAs (miRNA), and other signaling pathways, while the molecular mechanisms remain incompletely characterized. In this study, we used thoracic aortic constriction (TAC)-induced mice and angiotensin-II (Ang-II)-induced H9C2 cell line as cardiac hypertrophy model to investigate the role of miR-26a-5p in cardiac hypertrophy. We found that miR-26a-5p was downregulated in cardiac hypertrophy mice. Overexpression of miR-26a-5p by type 9 recombinant adeno-associated virus (rAAV9) reversed the heart hypertrophic manifestations. The phenotypes were also promoted by miR-26a-5p inhibitor in Ang-II-induced H9C2 cells. Through miRNA profile analysis and dual-luciferase reporter assay, ADAM17 was identified as a direct target of miR-26a-5p. Restored expression of ADAM17 disrupted the effect of miR-26a-5p on cardiac hypertrophy. To sum up, these results indicated that miR-26a-5p played an inhibitory role in cardiac hypertrophy and dysfunction via targeting ADAM17. The miR-26a-5p-ADAM17-cardiac hypertrophy axis provided special insight and a new molecular mechanism for a better understanding of cardiac hypertrophy disease, as well as the diagnostic and therapeutic practice.


Asunto(s)
Proteína ADAM17/metabolismo , Cardiomegalia/genética , MicroARNs/genética , Proteína ADAM17/genética , Angiotensina II/metabolismo , Animales , Autofagia/genética , Cardiomegalia/fisiopatología , Línea Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Transducción de Señal/genética
11.
Naunyn Schmiedebergs Arch Pharmacol ; 394(10): 2117-2128, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34398250

RESUMEN

The incidence of chronic kidney disease is escalating; cardiorenal syndrome (CRS) type 4 is gaining a major health concern causing significant morbidity and mortality, putting major burdens on the healthcare system. This study was designed to compare the cardioprotective effects of carvedilol versus atenolol against CRS type 4 induced by subtotal 5/6 nephrectomy in rats and to explore the underlying mechanisms. Immediately after surgery, carvedilol (20 mg/kg/day) or atenolol (20 mg/kg/day) was added to drinking water for 10 weeks. Carvedilol was more effective than atenolol in improving kidney functions, decreasing elevated blood pressures, attenuating cardiac hypertrophy, reducing serum brain natriuretic peptide, and diminished cardiac fibrous tissue deposition. However, carvedilol was equivalent to atenolol in modulating ß1-adrenergic receptors (ß1ARs) and cardiac diacylglycerol (DAG) signaling, but carvedilol was superior in modulating ß-arrestin2, phosphatidyl inositol 4,5 bisphosphates (PIP2), and caspase 3 levels. Carvedilol has superior cardioprotective effects than atenolol in a rat model of CRS type 4. These protective effects are mediated through modulating cardiac ß1ARs/ß-arrestin2/PIP2/DAG as well as abating cardiac apoptotic signaling pathways (caspase3/pS473 protein kinase B (Akt)).


Asunto(s)
Atenolol/uso terapéutico , Síndrome Cardiorrenal/tratamiento farmacológico , Cardiomegalia/tratamiento farmacológico , Cardiotónicos/uso terapéutico , Carvedilol/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Atenolol/farmacología , Presión Sanguínea/efectos de los fármacos , Síndrome Cardiorrenal/metabolismo , Síndrome Cardiorrenal/fisiopatología , Síndrome Cardiorrenal/cirugía , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Cardiomegalia/cirugía , Cardiotónicos/farmacología , Carvedilol/farmacología , Diacilglicerol Quinasa/metabolismo , Modelos Animales de Enfermedad , Riñón/efectos de los fármacos , Riñón/fisiología , Masculino , Miocardio/metabolismo , Nefrectomía , Fosfatidilinositol 4,5-Difosfato/metabolismo , Ratas Wistar , Receptores Adrenérgicos beta 1/metabolismo , Arrestina beta 2/metabolismo
12.
Clin Sci (Lond) ; 135(14): 1631-1647, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34296750

RESUMEN

Raf kinases signal via extracellular signal-regulated kinases 1/2 (ERK1/2) to drive cell division. Since activating mutations in BRAF (B-Raf proto-oncogene, serine/threonine kinase) are highly oncogenic, BRAF inhibitors including dabrafenib have been developed for cancer. Inhibitors of ERK1/2 signalling used for cancer are cardiotoxic in some patients, raising the question of whether dabrafenib is cardiotoxic. In the heart, ERK1/2 signalling promotes not only cardiomyocyte hypertrophy and is cardioprotective but also promotes fibrosis. Our hypothesis is that ERK1/2 signalling is not required in a non-stressed heart but is required for cardiac remodelling. Thus, dabrafenib may affect the heart in the context of, for example, hypertension. In experiments with cardiomyocytes, cardiac fibroblasts and perfused rat hearts, dabrafenib inhibited ERK1/2 signalling. We assessed the effects of dabrafenib (3 mg/kg/d) on male C57BL/6J mouse hearts in vivo. Dabrafenib alone had no overt effects on cardiac function/dimensions (assessed by echocardiography) or cardiac architecture. In mice treated with 0.8 mg/kg/d angiotensin II (AngII) to induce hypertension, dabrafenib inhibited ERK1/2 signalling and suppressed cardiac hypertrophy in both acute (up to 7 d) and chronic (28 d) settings, preserving ejection fraction. At the cellular level, dabrafenib inhibited AngII-induced cardiomyocyte hypertrophy, reduced expression of hypertrophic gene markers and almost completely eliminated the increase in cardiac fibrosis both in interstitial and perivascular regions. Dabrafenib is not overtly cardiotoxic. Moreover, it inhibits maladaptive hypertrophy resulting from AngII-induced hypertension. Thus, Raf is a potential therapeutic target for hypertensive heart disease and drugs such as dabrafenib, developed for cancer, may be used for this purpose.


Asunto(s)
Antineoplásicos/farmacología , Fibrosis/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Imidazoles/farmacología , Oximas/farmacología , Animales , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Hipertensión/fisiopatología , Ratones Endogámicos C57BL , Miocardio/patología , Miocitos Cardíacos/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
13.
JCI Insight ; 6(18)2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34324438

RESUMEN

Cardiac inflammation and fibrosis contribute significantly to hypertension-related adverse cardiac remodeling. IκB kinase ß (IKK-ß), a central coordinator of inflammation through activation of NF-κB, has been demonstrated as a key molecular link between inflammation and cardiovascular disease. However, the cell-specific contribution of IKK-ß signaling toward adverse cardiac remodeling remains elusive. Cardiac fibroblasts are one of the most populous nonmyocyte cell types in the heart that play a key role in mediating cardiac fibrosis and remodeling. To investigate the function of fibroblast IKK-ß, we generated inducible fibroblast-specific IKK-ß-deficient mice. Here, we report an important role of IKK-ß in the regulation of fibroblast functions and cardiac remodeling. Fibroblast-specific IKK-ß-deficient male mice were protected from angiotensin II-induced cardiac hypertrophy, fibrosis, and macrophage infiltration. Ablation of fibroblast IKK-ß inhibited angiotensin II-stimulated fibroblast proinflammatory and profibrogenic responses, leading to ameliorated cardiac remodeling and improved cardiac function in IKK-ß-deficient mice. Findings from this study establish fibroblast IKK-ß as a key factor regulating cardiac fibrosis and function in hypertension-related cardiac remodeling.


Asunto(s)
Angiotensina II/farmacología , Cardiomegalia/genética , Fibroblastos/fisiología , Quinasa I-kappa B/genética , Miocardio/patología , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colágeno Tipo I/metabolismo , Fibrosis , Técnicas de Silenciamiento del Gen , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/inducido químicamente , Inflamación/metabolismo , Macrófagos , Masculino , Ratones , Miocarditis/genética , Miocarditis/metabolismo , Tamaño de los Órganos , Factores Protectores , Transducción de Señal , Remodelación Ventricular/genética
14.
Benef Microbes ; 12(3): 283-293, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34030609

RESUMEN

Escherichia coli Nissle (EcN), a probiotic bacterium protects against several disorders. Multiple reports have studied the pathways involved in cardiac hypertrophy. However, the effects of probiotic EcN against diabetes-induced cardiac hypertrophy remain to be understood. We administered five weeks old Wistar male (271±19.4 g body weight) streptozotocin-induced diabetic rats with 109 cfu of EcN via oral gavage every day for 24 days followed by subjecting the rats to echocardiography to analyse the cardiac parameters. Overexpressed interleukin (IL)-6 induced the MEK5/ERK5, JAK2/STAT3, and MAPK signalling cascades in streptozotocin-induced diabetic rats. Further, the upregulation of calcineurin, NFATc3, and p-GATA4 led to the elevation of hypertrophy markers, such as atrial and B-type natriuretic peptides. In contrast, diabetic rats supplemented with probiotic EcN exhibited significant downregulated IL-6. Moreover, the MEK5/ERK5 and JAK2/STAT3 cascades involved during eccentric hypertrophy and MAPK signalling, including phosphorylated MEK, ERK, JNK, and p-38, were significantly attenuated in diabetic rats after supplementation of EcN. Western blotting and immunofluorescence revealed the significant downregulation of NFATc3 and downstream mediators, thereby resulting in the impairment of cardiac hypertrophy. Taken together, the findings demonstrate that supplementing probiotic EcN has the potential to show cardioprotective effects by inhibiting diabetes-induced cardiomyopathies.


Asunto(s)
Cardiomegalia/terapia , Diabetes Mellitus Experimental/terapia , Cardiomiopatías Diabéticas/terapia , Escherichia coli/fisiología , Interleucina-6/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Probióticos/uso terapéutico , Animales , Calcineurina/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , MAP Quinasa Quinasa 5/metabolismo , Masculino , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Ratas , Ratas Wistar , Factor de Transcripción STAT3/metabolismo , Estreptozocina
15.
J Cardiovasc Pharmacol ; 77(6): 822-829, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34016843

RESUMEN

ABSTRACT: In the previous study, we showed that an Hsp90 inhibitor, 17-(allylamino)-17-demethoxygeldanamycin (17-AAG), attenuates hypertrophic remodeling of cardiomyocytes during the development of heart failure. In this present study, we investigated the effects of 17-AAG on cardiac fibrosis during the development of heart failure. We used pressure-loaded cardiac hypertrophic mice prepared by constriction of the transverse aorta (TAC), which induces significant cardiac fibrosis without scar tissue. From the sixth week after the TAC operation, vehicle or 17-AAG was administered intraperitoneally twice a week. Eight weeks after the operation, the vehicle-treated animals showed chronic heart failure. On the other hand, cardiac deterioration of the 17-AAG-treated animals was attenuated. In 17-AAG-treated animals, when the degree of fibrosis was observed by histological staining, their volume of fibrosis was found to be reduced. The content of calcineurin, an Hsp90 client protein, and the level of dephosphorylated NFATc2, a transcription factor in the cardiac fibroblasts, in the TAC mice was reduced by treatment with 17-AAG. Furthermore, c-Raf and Erk signaling, indicators for cell proliferation and collagen synthesis, was also attenuated. In in vitro experiments, the proliferation and collagen synthesis of the cultured cardiac fibroblasts were attenuated by the presence of 17-AAG. When cardiac fibroblasts were incubated with angiotensin II, calcineurin-NFATc2 and c-Raf-Erk signaling in the cells were activated. These activations were attenuated by 17-AAG. Our findings suggest that suppression of the calcineurin-NFAT and c-Raf-Erk pathways may partially contribute to the attenuation of myocardial fibrosis caused by treatment with 17-AAG. Therefore, our data imply that the Hsp90 inhibitor may have potential for novel therapeutic strategy for the treatment of heart failure.


Asunto(s)
Benzoquinonas/farmacología , Cardiomegalia/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Lactamas Macrocíclicas/farmacología , Animales , Calcineurina/metabolismo , Cardiomegalia/fisiopatología , Proliferación Celular/efectos de los fármacos , Fibrosis/prevención & control , Insuficiencia Cardíaca/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Miocitos Cardíacos/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal/efectos de los fármacos
16.
J Cell Physiol ; 236(11): 7578-7590, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33928637

RESUMEN

Endothelial glycolytic metabolism plays an important role in the process of angiogenesis. TP53-induced glycolysis and apoptosis regulator (TIGAR) is a significant mediator of cellular energy homeostasis. However, the role of TIGAR in endothelial metabolism, angiogenesis, and coronary flow reserve (CFR) has not been studied. The present study investigated whether knockout (KO) of TIGAR improves endothelial glycolytic function and angiogenesis. In vitro, aortic endothelial cells (ECs) from TIGAR KO mice exhibited increased expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform-3 (PFKFB3) and increased glycolytic function. These were accompanied by increased mitochondrial basal/maximal respiration and ATP production. Furthermore, knockout of TIGAR in ECs enhanced endothelial proliferation, migration, and tube formation. Knockout of TIGAR also significantly increased aortic sprouting ex vivo. In vivo, knockout of TIGAR increased the expression of proangiogenic factor, angiopoietin-1 (Ang-1) in mouse hearts. Knockout of TIGAR also significantly increased coronary capillary density with enhanced CFR in these hearts. Furthermore, TIGAR KO mice subjected to pressure overload (PO), a common model to study angiogenesis and cardiac hypertrophy, exhibited elevated expression of Ang-1, VEGF, and PFKFB3 than that of the wild-type (WT) mice. WT mice subjected to PO exhibited a significant reduction of coronary capillary density and impaired CFR, but TIGAR KO mice did not. In addition, knockout of TIGAR blunted TAC-induced cardiac hypertrophy and dysfunction seen in the WT mice. In conclusion, knockout of TIGAR improves endothelial angiogenetic capabilities by enhancing the endothelial glycolytic function, mitochondrial respiration, and proangiogenic signaling, which leads to increased coronary capillary density and vascular function and protects against chronic stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Cardiomegalia/metabolismo , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Glucólisis , Neovascularización Fisiológica , Monoéster Fosfórico Hidrolasas/metabolismo , Angiopoyetina 1/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Circulación Coronaria , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Densidad Microvascular , Mitocondrias/genética , Mitocondrias/metabolismo , Fosfofructoquinasa-2/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda
17.
Cells ; 10(4)2021 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-33916597

RESUMEN

Cardiac remodeling and contractile dysfunction are leading causes in hypertrophy-associated heart failure (HF), increasing with a population's rising age. A hallmark of aged and diseased hearts is the accumulation of modified proteins caused by an impaired autophagy-lysosomal-pathway. Although, autophagy inducer rapamycin has been described to exert cardioprotective effects, it remains to be shown whether these effects can be attributed to improved cardiomyocyte autophagy and contractility. In vivo hypertrophy was induced by transverse aortic constriction (TAC), with mice receiving daily rapamycin injections beginning six weeks after surgery for four weeks. Echocardiographic analysis demonstrated TAC-induced HF and protein analyses showed abundance of modified proteins in TAC-hearts after 10 weeks, both reduced by rapamycin. In vitro, cardiomyocyte hypertrophy was mimicked by endothelin 1 (ET-1) and autophagy manipulated by silencing Atg5 in neonatal cardiomyocytes. ET-1 and siAtg5 decreased Atg5-Atg12 and LC3-II, increased natriuretic peptides, and decreased amplitude and early phase of contraction in cardiomyocytes, the latter two evaluated using ImageJ macro Myocyter recently developed by us. ET-1 further decreased cell contractility in control but not in siAtg5 cells. In conclusion, ET-1 decreased autophagy and cardiomyocyte contractility, in line with siAtg5-treated cells and the results of TAC-mice demonstrating a crucial role for autophagy in cardiomyocyte contractility and cardiac performance.


Asunto(s)
Autofagia , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Contracción Miocárdica , Miocardio/patología , Miocitos Cardíacos/patología , Animales , Animales Recién Nacidos , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Cardiomegalia/complicaciones , Cardiomegalia/diagnóstico por imagen , Ecocardiografía , Endotelina-1/metabolismo , Silenciador del Gen , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones Endogámicos C57BL , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Presión , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Disfunción Ventricular Izquierda/complicaciones , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/efectos de los fármacos
18.
Basic Res Cardiol ; 116(1): 26, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33876316

RESUMEN

Heart failure due to high blood pressure or ischemic injury remains a major problem for millions of patients worldwide. Despite enormous advances in deciphering the molecular mechanisms underlying heart failure progression, the cell-type specific adaptations and especially intercellular signaling remain poorly understood. Cardiac fibroblasts express high levels of cardiogenic transcription factors such as GATA-4 and GATA-6, but their role in fibroblasts during stress is not known. Here, we show that fibroblast GATA-4 and GATA-6 promote adaptive remodeling in pressure overload induced cardiac hypertrophy. Using a mouse model with specific single or double deletion of Gata4 and Gata6 in stress activated fibroblasts, we found a reduced myocardial capillarization in mice with Gata4/6 double deletion following pressure overload, while single deletion of Gata4 or Gata6 had no effect. Importantly, we confirmed the reduced angiogenic response using an in vitro co-culture system with Gata4/6 deleted cardiac fibroblasts and endothelial cells. A comprehensive RNA-sequencing analysis revealed an upregulation of anti-angiogenic genes upon Gata4/6 deletion in fibroblasts, and siRNA mediated downregulation of these genes restored endothelial cell growth. In conclusion, we identified a novel role for the cardiogenic transcription factors GATA-4 and GATA-6 in heart fibroblasts, where both proteins act in concert to promote myocardial capillarization and heart function by directing intercellular crosstalk.


Asunto(s)
Cardiomegalia/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Neovascularización Fisiológica , Remodelación Ventricular , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Aorta/fisiopatología , Aorta/cirugía , Presión Arterial , Cardiomegalia/etiología , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Comunicación Celular , Células Cultivadas , Constricción , Modelos Animales de Enfermedad , Fibroblastos/patología , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/genética , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones Noqueados , Densidad Microvascular , Miocardio/patología , Transducción de Señal
19.
Theranostics ; 11(10): 4710-4727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754023

RESUMEN

Background: Telomere shortening and dysfunction may cause metabolic disorders, tissue damage and age-dependent pathologies. However, little is known about the association of telomere-associated protein Rap1 with mitochondrial energy metabolism and cardiac aging. Methods: Echocardiography was performed to detect cardiac structure and function in Rap1+/+ and Rap1-/- mice at different ages (3 months, 12 months and 20 months). Telomere length, DNA damage, cardiac senescence and cardiomyocyte size were analyzed using the real-time PCR, Western blotting, senescence associated ß-galactosidase assay and wheat germ agglutinin staining, respectively. Western blotting was also used to determine the level of cardiac fatty acid metabolism related key enzymes in mouse and human myocardium. Chromatin immunoprecipitation assay was used to verify the direct link between p53 and PPARα. The p53 inhibitor, Pifithrin-α and PPARα activator WY14643 were utilized to identify the effects of Rap1/p53/PPARα signaling pathway. Results: Telomere was shortened concomitant with extensive DNA damage in aged Rap1-/- mouse hearts, evidenced by reduced T/S ratios and increased nuclear γH2AX. Meanwhile, the aging-associated phenotypes were pronounced as reflected by altered mitochondrial ultrastructure, enhanced senescence, cardiac hypertrophy and dysfunction. Mechanistically, acetylated p53 and nuclear p53 was enhanced in the Rap1-/- mouse hearts, concomitant with reduced PPARα. Importantly, p53 directly binds to the promoter of PPARα in mouse hearts and suppresses the transcription of PPARα. In addition, aged Rap1-/- mice exhibited reduced cardiac fatty acid metabolism. Pifithrin-α alleviated cardiac aging and enhanced fatty acid metabolism in the aged Rap1-/- mice. Activating PPARα with WY14643 in primarily cultured Rap1-/- cardiomyocytes restored maximal oxygen consumption rates. Reduced Rap1 expression and impaired p53/PPARα signaling also presented in aged human myocardium. Conclusion: In summary, Rap1 may link telomere biology to fatty acid metabolism and aging-related cardiac pathologies via modulating the p53/PPARα signaling pathway, which could represent a therapeutic target in preventing/attenuating cardiac aging.


Asunto(s)
Envejecimiento/genética , Cardiomegalia/genética , Senescencia Celular/genética , Miocitos Cardíacos/metabolismo , PPAR alfa/genética , Proteínas de Unión a Telómeros/genética , Proteína p53 Supresora de Tumor/genética , Animales , Benzotiazoles/farmacología , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/fisiopatología , Daño del ADN , Ecocardiografía , Ácidos Grasos/metabolismo , Cardiopatías/diagnóstico por imagen , Cardiopatías/genética , Cardiopatías/fisiopatología , Histonas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/ultraestructura , Prueba de Campo Abierto , PPAR alfa/metabolismo , Proliferadores de Peroxisomas/farmacología , Pirimidinas/farmacología , Complejo Shelterina , Transducción de Señal , Telómero/metabolismo , Homeostasis del Telómero , Proteínas de Unión a Telómeros/metabolismo , Tolueno/análogos & derivados , Tolueno/farmacología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo
20.
Theranostics ; 11(8): 3830-3838, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33664864

RESUMEN

Anti-angiogenics drugs in clinical use for cancer treatment induce cardiotoxic side effects. The endothelin axis is involved in hypertension and cardiac remodelling, and addition of an endothelin receptor antagonist to the anti-angiogenic sunitinib was shown to reduce cardiotoxicity of sunitinib in mice. Here, we explored further the antidote effect of the endothelin receptor antagonist macitentan in sunitinib-treated animals on cardiac remodeling. Methods: Tumor-bearing mice treated per os daily by sunitinib or vehicle were imaged before and after 1, 3 and 6 weeks of treatment by positron emission tomography using [18F]fluorodeoxyglucose and by echocardiography. Non-tumor-bearing animals were randomly assigned to be treated per os daily by vehicle or sunitinib or macitentan or sunitinib+macitentan, and imaged by echocardiography after 5 weeks. Hearts were harvested for histology and molecular analysis at the end of in vivo exploration. Results: Sunitinib treatment increases left ventricular mass and ejection fraction and induces cardiac fibrosis. Sunitinib also induces an early increase in cardiac uptake of [18F]fluorodeoxyglucose, which is significantly correlated with increased left ventricular mass at the end of treatment. Co-administration of macitentan prevents sunitinib-induced hypertension, increase in ejection fraction and cardiac fibrosis, but fails to prevent increase of the left ventricular mass. Conclusion: Early metabolic changes predict sunitinib-induced cardiac remodeling. Endothelin blockade can prevent some but not all cardiotoxic side-effects of sunitinib, in particular left ventricle hypertrophy that appears to be induced by sunitinib through an endothelin-independent mechanism.


Asunto(s)
Cardiomegalia/inducido químicamente , Endotelinas/fisiología , Sunitinib/toxicidad , Animales , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Antagonistas de los Receptores de Endotelina/administración & dosificación , Femenino , Fibrosis , Glucólisis/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Medicina de Precisión , Pirimidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA