Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
Mol Ther ; 29(3): 1120-1137, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33130312

RESUMEN

Emerging evidence reveals that autophagy plays crucial roles in cardiac hypertrophy. Long noncoding RNAs (lncRNAs) are novel transcripts that function as gene regulators. However, it is unclear whether lncRNAs regulate autophagy in cardiac hypertrophy. Here, we identified a novel transcript named lncRNA Gm15834, which was upregulated in the transverse aortic constriction (TAC) model in vivo and the angiotensin-II (Ang-II)-induced cardiac hypertrophy model in vitro and was regulated by nuclear factor kappa B (NF-κB). Importantly, forced expression of lncRNA Gm15834 enhanced autophagic activity of cardiomyocytes and promoted myocardial hypertrophy, whereas silencing of lncRNA Gm15834 attenuated autophagy-induced myocardial hypertrophy. Mechanistically, we found that lncRNA Gm15834 could function as an endogenous sponge RNA of microRNA (miR)-30b-3p, which was downregulated in cardiac hypertrophy. Inhibition of miR-30b-3p enhanced cardiomyocyte autophagic activity and aggravated myocardial hypertrophy, whereas overexpression of miR-30b-3p suppressed autophagy-induced myocardial hypertrophy by targeting the downstream autophagy factor of unc-51-like kinase 1 (ULK1). Moreover, inhibition of lncRNA Gm15834 by adeno-associated virus carrying short hairpin RNA (shRNA) suppressed cardiomyocyte autophagic activity, improved cardiac function, and mitigated cardiac hypertrophy. Taken together, our study identified a novel regulatory axis encompassing lncRNA Gm15834/miR-30b-3p/ULK1/autophagy in cardiac hypertrophy, which may provide a potential therapy target for cardiac hypertrophy.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia , Cardiomegalia/terapia , Regulación de la Expresión Génica , ARN Largo no Codificante/antagonistas & inhibidores , Angiotensina II/toxicidad , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal , Vasoconstrictores/toxicidad
2.
J Cell Mol Med ; 25(9): 4408-4419, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33793066

RESUMEN

Nuclear factor erythroid 2-related factor (Nrf2) is an important regulator of cellular antioxidant defence. We previously showed that SFN prevented Ang II-induced cardiac damage via activation of Nrf2. However, the underlying mechanism of SFN's persistent cardiac protection remains unclear. This study aimed to explore the potential of SFN in activating cardiac Nrf2 through epigenetic mechanisms. Wild-type mice were injected subcutaneously with Ang II, with or without SFN. Administration of chronic Ang II-induced cardiac inflammatory factor expression, oxidative damage, fibrosis and cardiac remodelling and dysfunction, all of which were effectively improved by SFN treatment, coupled with an up-regulation of Nrf2 and downstream genes. Bisulfite genome sequencing and chromatin immunoprecipitation (ChIP) were performed to detect the methylation level of the first 15 CpGs and histone H3 acetylation (Ac-H3) status in the Nrf2 promoter region, respectively. The results showed that SFN reduced Ang II-induced CpG hypermethylation and promoted Ac-H3 accumulation in the Nrf2 promoter region, accompanied by the inhibition of global DNMT and HDAC activity, and a decreased protein expression of key DNMT and HDAC enzymes. Taken together, SFN exerts its cardioprotective effect through epigenetic modification of Nrf2, which may partially contribute to long-term activation of cardiac Nrf2.


Asunto(s)
Angiotensina II/toxicidad , Cardiomiopatías/prevención & control , Epigénesis Genética , Regulación de la Expresión Génica/efectos de los fármacos , Isotiocianatos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Sulfóxidos/farmacología , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Vasoconstrictores/toxicidad
3.
J Cell Mol Med ; 25(16): 7760-7771, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34180125

RESUMEN

Lymphangiogenesis is possibly capable of attenuating hypertension-induced cardiac injury. Sirtuin 3 (SIRT3) is an effective mitochondrial deacetylase that has the potential to modulate this process; however, its role in hypertension-induced cardiac lymphangiogenesis to date has not been investigated. Our experiments were performed on 8-week-old wild-type (WT), SIRT3 knockout (SIRT3-KO) and SIRT3 overexpression (SIRT3-LV) mice infused with angiotensin II (Ang II) (1000 ng/kg per minute) or saline for 28 days. After Ang II infusion, SIRT3-KO mice developed a more severe cardiac remodelling, less lymphatic capillaries and lower expression of lymphatic marker when compared to wild-type mice. In comparison, SIRT3-LV restored lymphangiogenesis and attenuated cardiac injury. Furthermore, lymphatic endothelial cells (LECs) exposed to Ang II in vitro exhibited decreased migration and proliferation. Silencing SIRT3 induced functional decrease in LECs, while SIRT3 overexpression LECs facilitated. Moreover, SIRT3 may up-regulate lymphangiogenesis by affecting vascular endothelial growth factor receptor 3 (VEGFR3) and ERK pathway. These findings suggest that SIRT3 could promote lymphangiogenesis and attenuate hypertensive cardiac injury.


Asunto(s)
Angiotensina II/toxicidad , Células Endoteliales/patología , Lesiones Cardíacas/patología , Hipertensión/complicaciones , Linfangiogénesis , Sirtuina 3/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Lesiones Cardíacas/etiología , Lesiones Cardíacas/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Noqueados , Sirtuina 3/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Vasoconstrictores/toxicidad
4.
Neuroimage ; 232: 117919, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33652141

RESUMEN

Unilateral damage to the frontoparietal network typically impairs saccade target selection within the contralesional visual hemifield. Severity of deficits and the degree of recovery have been associated with widespread network dysfunction, yet it is not clear how these behavioural and functional brain changes relate with the underlying structural white matter tracts. Here, we investigated whether recovery after unilateral prefrontal cortex (PFC) lesions was associated with changes in white matter microstructure across large-scale frontoparietal cortical and thalamocortical networks. Diffusion-weighted imaging was acquired in four male rhesus macaques at pre-lesion, week 1, and week 8-16 post-lesion when target selection deficits largely recovered. Probabilistic tractography was used to reconstruct cortical frontoparietal fiber tracts, including the superior longitudinal fasciculus (SLF) and transcallosal fibers connecting the PFC or posterior parietal cortex (PPC), as well as thalamocortical fiber tracts connecting the PFC and PPC to thalamic nuclei. We found that the two animals with small PFC lesions showed increased fractional anisotropy in both cortical and thalamocortical fiber tracts when behaviour had recovered. However, we found that fractional anisotropy decreased in cortical frontoparietal tracts after larger PFC lesions yet increased in some thalamocortical tracts at the time of behavioural recovery. These findings indicate that behavioural recovery after small PFC lesions may be supported by both cortical and subcortical areas, whereas larger PFC lesions may have induced widespread structural damage and hindered compensatory remodeling in the cortical frontoparietal network.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/métodos , Red Nerviosa/diagnóstico por imagen , Corteza Prefrontal/diagnóstico por imagen , Recuperación de la Función/fisiología , Tálamo/diagnóstico por imagen , Sustancia Blanca/diagnóstico por imagen , Animales , Macaca mulatta , Masculino , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Estimulación Luminosa/métodos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Recuperación de la Función/efectos de los fármacos , Tálamo/efectos de los fármacos , Tálamo/fisiología , Vasoconstrictores/toxicidad , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/fisiología
5.
Amino Acids ; 53(7): 1079-1089, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34089389

RESUMEN

The present study was to explore whether alarin could alleviate heart failure (HF) and attenuate cardia fibrosis via inhibiting oxidative stress. The fibrosis of cardiac fibroblasts (CFs) was induced by angiotensin (Ang) II. HF models were induced by ligation of the left anterior descending artery to cause ischemia myocardial infarction (MI) in Sprague-Dawley rats. Alarin (1.0 nM/kg/d) was administrated by intraperitoneal injection for 28 days. The decreases of left ventricular (LV) ejection fraction (EF), fractional shortening (FS), the maximum of the first differentiation of LV pressure (LV ± dp/dtmax) and LV systolic pressure (LVSP), and the increases of LV volume in systole (LVVS), LV volume in diastole (LVVD), LV end-systolic diameter (LVESD) and LV end-diastolic diameter (LVEDD) in MI rats were improved by alarin treatment. The increases in the expression levels of collagen I, collagen III, and transforming growth factor (TGF)-ß were inhibited by alarin treatment in CFs and in the hearts of MI rats. The levels of NADPH oxidase (Nox) activity, superoxide anions and malondialdehyde (MDA) levels were increased, and the level of superoxide dismutase (SOD) activity was reduced in Ang II-treated CFs, which were reversed by alarin. Nox1 overexpression reversed the effects of alarin on attenuating the increases of collagen I, collagen III and TGF-ß expression levels induced by Ang II in CFs. These results indicated that alarin improved HF and cardiac fibrosis via inhibiting oxidative stress in HF rats. Nox1 played important roles in the regulation of alarin effects on attenuating CFs fibrosis induced by Ang II.


Asunto(s)
Angiotensina II/toxicidad , Fibrosis/prevención & control , Péptido Similar a Galanina/farmacología , Insuficiencia Cardíaca/complicaciones , Infarto del Miocardio/complicaciones , Estrés Oxidativo , Animales , Fibrosis/etiología , Fibrosis/patología , Masculino , Malondialdehído/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/metabolismo , Vasoconstrictores/toxicidad
6.
Mol Cell Biochem ; 476(9): 3253-3260, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33886061

RESUMEN

Pathological cardiac hypertrophy is associated with many diseases including hypertension. Recent studies have identified important roles for microRNAs (miRNAs) in many cardiac pathophysiological processes, including the regulation of cardiomyocyte hypertrophy. However, the role of miR-145-5p in the cardiac setting is still unclear. In this study, H9C2 cells were overexpressed with microRNA-145-5p, and then treated with Ang-II for 24 h, to study the effect of miR-145-5p on Ang-II-induced myocardial hypertrophy in vitro. Results showed that Ang-II treatment down-regulated miR-145-5p expression were revered after miR-145-5p overexpression. Based on results of bioinformatics algorithms, paxillin was predicted as a candidate target gene of miR-145-5p, luciferase activity assay revealed that the luciferase activity of cells was substantial downregulated the following co-transfection with wild paxillin 3'UTR and miR-145-5p compared to that in scramble control, while the inhibitory effect of miR-145-5p was abolished after transfection of mutant paxillin 3'UTR. Additionally, overexpression of miR-145-5p markedly inhibited activation of Rac-1/ JNK /c-jun/ NFATc3 and ANP expression and induced SIRT1 expression in Ang-II treated H9c2 cells. Jointly, our study suggested that miR-145-5p inhibited cardiac hypertrophy by targeting paxillin and through modulating Rac-1/ JNK /c-jun/ NFATc3/ ANP / Sirt1 signaling, therefore proving novel downstream molecular pathway of miR-145-5p in cardiac hypertrophy.


Asunto(s)
Angiotensina II/toxicidad , Cardiomegalia/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Mioblastos Cardíacos/efectos de los fármacos , Paxillin/antagonistas & inhibidores , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Cardiomegalia/patología , Células Cultivadas , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Sirtuina 1/genética , Sirtuina 1/metabolismo , Vasoconstrictores/toxicidad , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
7.
Can J Physiol Pharmacol ; 99(10): 1036-1047, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33857387

RESUMEN

Although vasodilatation evoked by acidosis at normal body temperature is well known, the reports regarding effect of acidosis on the reactivity of the isolated arteries at low temperatures are nonexistent. This study tested the hypothesis that the inhibitory effect of acidosis on the neurogenic vasoconstriction may be increased by cooling. Using wire myography, we recorded the neurogenic contraction of the rat tail artery segments to the electrical field stimulation in the absence and in the presence of 0.03-10.0 µmol/L noradrenaline. The experiments were conducted at 37 °C or 25 °C and pH 7.4 or 6.6 which was decreased by means of CO2. Noradrenaline at concentration of 0.03-0.1 µmol/L significantly potentiated the neurogenic vasoconstriction at 25 °C, and the potentiation was not inhibited by acidosis. Contrary to our hypothesis, acidosis at a low temperature did not affect the noradrenaline-induced tone and significantly increased the neurogenic contraction of the artery segments in the absence and presence of noradrenaline. These effects of acidosis were partly dependent on the endothelium and L-type Ca2+ channels activation. The phenomenon described for the first time might be of importance for the reduction in the heat loss by virtue of decrease in the subcutaneous blood flow at low ambient temperatures.


Asunto(s)
Acidosis/fisiopatología , Norepinefrina/farmacología , Nervios Periféricos/patología , Animales , Frío , Estimulación Eléctrica , Masculino , Contracción Muscular , Óxido Nítrico/metabolismo , Nervios Periféricos/efectos de los fármacos , Ratas , Ratas Wistar , Cola (estructura animal)/irrigación sanguínea , Vasoconstricción , Vasoconstrictores/toxicidad
8.
Can J Physiol Pharmacol ; 99(10): 1000-1006, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33852804

RESUMEN

The Hippo/YAP (yes-associated protein) pathway is an important signaling pathway to control organ development and tissue homeostasis. YAP is a downstream effector of the Hippo pathway and a critical mediator of mechanic stress. Hypertensive nephropathy is characterized with glomerular sclerosis stiffness and renal fibrosis. The present study investigated the role of YAP pathway in angiotensin (Ang) II hypertensive renal injury by using YAP activation inhibitor verteporfin. Ang II increased the protein expression of YAP in renal nucleus fraction, decreased phospho-YAP, and phospho-LATS1/2 (large tumor suppressors 1 and 2) expressions in renal cytoplasmic fraction, suggesting Ang II activation of renal YAP. Ang II significantly increased systolic blood pressure (SBP), proteinuria, glomerular sclerosis, and fibrosis; treatment with verteporfin attenuated Ang II-induced proteinuria and renal injury with a mild reduction in SBP. Moreover, Ang II increased the protein expressions of inflammatory factors including tumor necrosis factor α, interleukin 1ß, and monocyte chemoattractant protein-1, and profibrotic factors including transforming growth factor ß, phospho-Smad3 and fibronectin. Verteporfin reversed abovementioned Ang II-induced molecule expressions. Our results for the first time demonstrate that the activation of the YAP pathway promotes hypertensive renal inflammation and fibrosis, which may promote hypertensive renal injury. YAP may be a new target for prevention and treatment of hypertensive renal diseases.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Angiotensina II/toxicidad , Hipertensión Renal/tratamiento farmacológico , Hipertensión/metabolismo , Nefritis/tratamiento farmacológico , Verteporfina/farmacología , Proteínas Señalizadoras YAP/antagonistas & inhibidores , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Presión Sanguínea , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Fibrosis , Hipertensión/inducido químicamente , Hipertensión/patología , Hipertensión Renal/etiología , Hipertensión Renal/metabolismo , Hipertensión Renal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Nefritis/etiología , Nefritis/metabolismo , Nefritis/patología , Fármacos Fotosensibilizantes/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Vasoconstrictores/toxicidad
9.
Int J Mol Sci ; 22(5)2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33668403

RESUMEN

Cocaine is one of the most widely abused illicit drugs worldwide and has long been recognised as an agent of cardiac dysfunction in numerous cases of drug overdose. Cocaine has previously been shown to up-regulate cytoskeletal rearrangements and morphological changes in numerous tissues; however, previous literature observes such changes primarily in clinical case reports and addiction studies. An investigation into the fundamental cytoskeletal parameters of migration, adhesion and proliferation were studied to determine the cytoskeletal and cytotoxic basis of cocaine in cardiac cells. Treatment of cardiac myocytes with cocaine increased cell migration and adhesion (p < 0.05), with no effect on cell proliferation, except with higher doses eliciting (1-10 µg/mL) its diminution and increase in cell death. Cocaine downregulated phosphorylation of cofilin, decreased expression of adhesion modulators (integrin-ß3) and increased expression of ezirin within three hours of 1 µg/mL treatments. These functional responses were associated with changes in cellular morphology, including alterations in membrane stability and a stellate-like phenotype with less compaction between cells. Higher dose treatments of cocaine (5-10 µg/mL) were associated with significant cardiomyocyte cell death (p < 0.05) and loss of cellular architecture. These results highlight the importance of cocaine in mediating cardiomyocyte function and cytotoxicity associated with the possible loss of intercellular contacts required to maintain normal cell viability, with implications for cardiotoxicity relating to hypertrophy and fibrogenesis.


Asunto(s)
Cardiotoxicidad/patología , Cocaína/toxicidad , Citoesqueleto/patología , Miocitos Cardíacos/patología , Vasoconstrictores/toxicidad , Animales , Cardiotoxicidad/etiología , Adhesión Celular , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Ratas
10.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33947043

RESUMEN

Neonatal arterial ischemic stroke is one of the more severe birth complications. The injury can result in extensive neurological damage and is robustly associated with later diagnoses of cerebral palsy (CP). An important part of efforts to develop new therapies include the on-going refinement and understanding of animal models that capture relevant clinical features of neonatal brain injury leading to CP. The potent vasoconstrictor peptide, Endothelin-1 (ET-1), has previously been utilised in animal models to reduce local blood flow to levels that mimic ischemic stroke. Our previous work in this area has shown that it is an effective and technically simple approach for modelling ischemic injury at very early neonatal ages, resulting in stable deficits in motor function. Here, we aimed to extend this model to also examine the impact on cognitive function. We show that focal delivery of ET-1 to the cortex of Sprague Dawley rats on postnatal day 0 (P0) resulted in impaired learning in a touchscreen-based test of visual discrimination and correlated with important clinical features of CP including damage to large white matter structures.


Asunto(s)
Isquemia Encefálica/complicaciones , Parálisis Cerebral/etiología , Modelos Animales de Enfermedad , Endotelina-1/toxicidad , Vasoconstrictores/toxicidad , Animales , Animales Recién Nacidos , Aprendizaje por Asociación , Atrofia , Isquemia Encefálica/inducido químicamente , Isquemia Encefálica/patología , Recuento de Células , Corteza Cerebral/patología , Parálisis Cerebral/patología , Trastornos del Conocimiento/etiología , Cuerpo Estriado/patología , Endotelina-1/administración & dosificación , Inflamación , Inyecciones , Microglía/patología , Trastornos del Movimiento/etiología , Neuronas/patología , Trastornos de la Percepción/etiología , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante , Vasoconstrictores/administración & dosificación , Sustancia Blanca/patología
11.
Int J Mol Sci ; 22(10)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064664

RESUMEN

Rutin is a flavonoid with antioxidant property. It has been shown to exert cardioprotection against cardiomyocyte hypertrophy. However, studies regarding its antihypertrophic property are still lacking, whether it demonstrates similar antihypertrophic effect to its metabolite, quercetin. Hence, this study aimed to investigate the effects of both flavonoids on oxidative stress and mitogen-activated protein kinase (MAPK) pathway in H9c2 cardiomyocytes that were exposed to angiotensin II (Ang II) to induce hypertrophy. Cardiomyocytes were exposed to Ang II (600 nM) with or without quercetin (331 µM) or rutin (50 µM) for 24 h. A group given vehicle served as the control. The concentration of the flavonoids was chosen based on the reported effective concentration to reduce cell hypertrophy or cardiac injury in H9c2 cells. Exposure to Ang II increased cell surface area, intracellular superoxide anion level, NADPH oxidase and inducible nitric oxide synthase activities, and reduced cellular superoxide dismutase activity and nitrite level, which were similarly reversed by both rutin and quercetin. Rutin had no significant effects on phosphorylated proteins of extracellular signal-related kinases (ERK1/2) and p38 but downregulated phosphorylated c-Jun N-terminal kinases (JNK1/2), which were induced by Ang II. Quercetin, on the other hand, had significantly downregulated the phosphorylated proteins of ERK1/2, p38, and JNK1/2. The quercetin inhibitory effect on JNK1/2 was stronger than the rutin. In conclusion, both flavonoids afford similar protective effects against Ang II-induced cardiomyocyte hypertrophy, but they differently modulate MAPK pathway.


Asunto(s)
Angiotensina II/toxicidad , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hipertrofia/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mioblastos Cardíacos/metabolismo , Quercetina/farmacología , Rutina/farmacología , Animales , Antioxidantes/farmacología , Células Cultivadas , Hipertrofia/inducido químicamente , Hipertrofia/tratamiento farmacológico , Hipertrofia/patología , Proteínas Quinasas Activadas por Mitógenos/genética , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/efectos de los fármacos , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Fosforilación , Ratas , Especies Reactivas de Oxígeno/metabolismo , Vasoconstrictores/toxicidad
12.
Am J Physiol Cell Physiol ; 318(3): C555-C569, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31940248

RESUMEN

Unlike the pulmonary artery (PA), the pathophysiological changes of the pulmonary vein (PV) in the development of pulmonary hypertension (PH) remain largely unknown. In this study, we comprehensively investigated the structural and functional changes in the PV isolated from the chronic hypoxia (CH; 10% O2, 21 days)-induced PH rat model (CHPH). Results showed that CH caused an increase in right ventricular pressure but did not affect the mean pulmonary venous pressure and the left atrial pressure. Similar to the PA, vascular lumen stenosis and medial thickening were also observed in the intrapulmonary veins isolated from the CHPH rats. Notably, CH induced more severe loss in the endothelium of intrapulmonary veins than the arteries. Then, the contractile response to 5-HT and U46619 was significantly greater in the intrapulmonary small veins (ISPV) and arteries (ISPA) isolated from CHPH rats than those from normoxic rats but not in the extrapulmonary and intrapulmonary large veins. Treatment with nifedipine (Nif), SKF96365 (SKF), or ryanodine and caffeine either partially attenuated (Nif) or dramatically abolished (SKF or ryanodine and caffeine) 5-HT-induced maximal contraction in ISPV from both normoxic and CHPH rats. Because of the severe loss of endothelium in the PV of CHPH rats, the decrease in acetylcholine (ACh)-induced endothelium-dependent relaxation was significantly larger in ISPV than ISPA, whereas the sodium nitroprusside-induced endothelium-independent relaxation was not altered in both ISPA and ISPV. In conclusion, our results provide fundamental data to comprehensively define the PV system in CHPH rat model.


Asunto(s)
Modelos Animales de Enfermedad , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Venas Pulmonares/citología , Venas Pulmonares/fisiología , Animales , Células Cultivadas , Enfermedad Crónica , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/patología , Hipoxia/patología , Masculino , Técnicas de Cultivo de Órganos , Venas Pulmonares/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Vasoconstrictores/toxicidad , Vasodilatadores/farmacología
13.
J Recept Signal Transduct Res ; 40(6): 493-500, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32496870

RESUMEN

Cardiac fibrosis is a pathological feature common to a variety of heart diseases such as myocardial infarction, arrhythmias, cardiomyopathies and heart failure. The molecular mechanism underlying the cardiac fibrosis is still unclear. Forkhead box F1 (FOXF1), a member of the forkhead transcription factor superfamily, plays critical roles in the development of hepatic fibrosis. However, whether FOXF1 is involved in the pathogenesis of cardiac fibrosis remains to be elucidated. The present study aimed to investigate the role of FOXF1 and its mechanisms in regulating cardiac fibrosis. The results demonstrated that FOXF1 was downregulated in Ang II-induced CFs. Overexpression of FOXF1 inhibited angiotensin II (Ang II)-induced proliferation, migration and oxidative stress in cardiac fibroblasts (CFs). Overexpression of FOXF1 also reduced the expression of alpha-smooth muscle actin (a-SMA) in Ang II-induced CFs, suggesting that overexpression of FOXF1 prevented the differentiation of CFs to myofibroblasts. Furthermore, the production of extracellular matrix (ECM) components including type I collagen and fibronectin were reduced by overexpression of FOXF1 in Ang II-induced CFs. Furthermore, overexpression of FOXF1 prevented Ang II-induced activation of transforming growth factor beta 1 (TGF-ß1)/Smad3 pathway in CFs. In conclusion, the results of the present study indicated that FOXF1 acted as a key regulator of pathological cardiac fibrosis, and overexpression of FOXF1 ameliorated cardiac fibrosis by inhabiting the TGF-ß1/Smad3 signaling pathway. These results indicated that FOXF1 may be a novel target for attenuating cardiac fibrosis.


Asunto(s)
Angiotensina II/toxicidad , Fibrosis/prevención & control , Factores de Transcripción Forkhead/metabolismo , Cardiopatías/prevención & control , Miofibroblastos/efectos de los fármacos , Proteína smad3/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Proliferación Celular , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Fibrosis/patología , Factores de Transcripción Forkhead/genética , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Cardiopatías/patología , Masculino , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Vasoconstrictores/toxicidad
14.
FASEB J ; 33(1): 494-500, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30118322

RESUMEN

Dimethylarginine dimethylaminohydrolases (DDAHs) are known to degrade asymmetric dimethylarginine, an endogenous inhibitor of NOS, and maintain vascular homeostasis; however, the regulatory pathways of DDAHs remain unclear. In this study, we aimed to define the role of transmembrane glycoprotein neuropilin-1 (NRP1) in the expression of DDAHs and investigate the potential roles of NRP1 in regulation of blood pressure. Short hairpin RNA-mediated knockdown of NRP1 reduced the level and mRNA stability of DDAH1 but not DDAH2 in HUVECs, whereas overexpression of NRP1 increased the mRNA stability of DDAH1. Meanwhile, mesenteric arteries and lung vascular endothelial cells of tamoxifen-inducible endothelial cell-specific NRP1 knockout mice exhibited decreased expression of DDAH1 and slightly increased expression of DDAH2. Mechanistically, the regulation of NRP1 on DDAH1 expression is mediated by a posttranscriptional mechanism involving miR-219-5p in HUVECs. Although the endothelial cell-specific NRP1 knockout mice did not exhibit any significant change in blood pressure at the basal level, they were more sensitive to low-dose angiotensin II infusion-induced increases in blood pressure. Our results show that NRP1 is required for full expression of DDAH1 in endothelial cells and that NRP1 contributes to protection from low-dose angiotensin II-induced increases in blood pressure.-Wang, Y., Wang, E., Zhang, Y., Madamsetty, V. S., Ji, B., Radisky, D. C., Grande, J. P., Misra, S., Mukhopadhyay, D. Neuropilin-1 maintains dimethylarginine dimethylaminohydrolase 1 expression in endothelial cells, and contributes to protection from angiotensin II-induced hypertension.


Asunto(s)
Amidohidrolasas/fisiología , Angiotensina II/toxicidad , Endotelio Vascular/efectos de los fármacos , Hipertensión/prevención & control , Neuropilina-1/fisiología , Vasoconstrictores/toxicidad , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Presión Sanguínea , Células Cultivadas , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo
15.
Nephrol Dial Transplant ; 35(9): 1491-1500, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32500132

RESUMEN

BACKGROUND: We have shown that the CXCL16/CXCR6 axis plays a critical role in recruiting inflammatory cells and bone marrow-derived fibroblasts into the kidney leading to renal injury and fibrosis. However, the underlying signaling mechanisms are not known. METHODS: In the present study, we examined the role of phosphoinositide-3 kinase γ (PI3Kγ) signaling in the recruitment of inflammatory cells and bone marrow-derived fibroblasts into the kidney and development of renal injury and fibrosis in an experimental model of hypertension induced by angiotensin II. RESULTS: Blood pressure was comparable between wild-type (WT) and PI3Kγ knockout (KO) mice at baseline. Angiotensin II treatment led to an increase in blood pressure that was similar between WT and PI3Kγ KO mice. Compared with WT mice, PI3Kγ KO mice were protected from angiotensin II-induced renal dysfunction and injury and developed less proteinuria. PI3Kγ deficiency suppressed bone marrow-derived fibroblast accumulation and myofibroblast formation in the kidney and inhibited total collagen deposition and extracellular matrix protein production in the kidney in response to angiotensin II. PI3Kγ deficiency inhibited the infiltration of F4/80+ macrophages and CD3+ T cells into the kidney and reduced gene expression levels of pro-inflammatory cytokines in the kidney following angiotensin II treatment. Finally, inhibition of PI3Kγ suppressed CXCL16-induced monocyte migration in vitro. CONCLUSION: These results indicate that PI3Kγ mediates the influx of macrophages, T cells and bone marrow-derived fibroblasts into the kidney resulting in kidney injury and fibrosis.


Asunto(s)
Lesión Renal Aguda/prevención & control , Angiotensina II/toxicidad , Fosfatidilinositol 3-Quinasa Clase Ib/fisiología , Fibrosis/prevención & control , Hipertensión/complicaciones , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Hipertensión/inducido químicamente , Hipertensión/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Miofibroblastos/patología , Linfocitos T/metabolismo , Linfocitos T/patología , Vasoconstrictores/toxicidad
16.
J Mol Cell Cardiol ; 132: 49-59, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31071332

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease without an effective pharmaceutical treatment. Genetic studies have proved the involvement of smooth muscle phenotype switch in the development of AAA. The alpha subunit of the heterotrimeric G stimulatory protein (Gsα) mediates receptor-stimulated production of cyclic adenosine monophosphate (cAMP). However, the role of smooth muscle Gsα in AAA formation remains unknown. APPROACH AND RESULTS: In this study, mice with knockout of smooth muscle-specific Gsα (GsαSMKO) were generated by cross-breeding Gsαflox/flox mice with SM22-CreERT2 transgenic mice, induced in adult mice by tamoxifen treatment. Gsα deficiency induced a smooth muscle phenotype switch from a contractile to a synthetic state. Mechanically, Gsα deletion reduced cAMP level and increased the level of human antigen R (HuR), which binds with the adenylate uridylate-rich elements of the 3' untranslated region of Krüppel-like factor 4 (KLF4) mRNA, thereby increasing the stability of KLF4. Moreover, genetic knockdown of HuR or KLF4 rescued the phenotype switch in Gsα-deficient smooth muscle cells. Furthermore, with acute infusion of angiotensin II, the incidence of AAA was markedly higher in ApoE-/-/GsαSMKO than ApoE-/-/Gsαflox/flox mice and induced increased elastic lamina degradation and aortic expansion. Finally, the levels of Gsα and SM α-actin were significantly lower while those of HuR and KLF4 were higher in human AAA samples than adjacent nonaneurysmal aortic sections. CONCLUSIONS: Gsα may play a protective role in AAA formation by regulating the smooth muscle phenotype switch and could be a potential therapeutic target for AAA disease.


Asunto(s)
Angiotensina II/toxicidad , Aneurisma de la Aorta Abdominal/etiología , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Músculo Liso Vascular/patología , Vasoconstrictores/toxicidad , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Modelos Animales de Enfermedad , Humanos , Factor 4 Similar a Kruppel , Masculino , Ratones , Ratones Noqueados para ApoE , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Fenotipo , Transducción de Señal
17.
Bioorg Med Chem Lett ; 29(19): 126616, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31439380

RESUMEN

19-Hydroxyeicosatetraenoic acid (19-HETE, 1), a metabolically and chemically labile cytochrome P450 eicosanoid, has diverse biological activities including antagonism of the vasoconstrictor 20-hydroxyeicosatetraenoic acid (20-HETE, 2). A SAR study was conducted to develop robust analogs of 1 with improved in vitro and in vivo efficacy. Analogs were screened in vitro for inhibition of 20-HETE-induced sensitization of rat renal preglomerular microvessels toward phenylephrine and demonstrated to normalize the blood pressure of male Cyp4a14(-/-) mice that display androgen-driven, 20-HETE-dependent hypertension.


Asunto(s)
Antihipertensivos/química , Antihipertensivos/farmacología , Familia 4 del Citocromo P450/fisiología , Hipertensión/tratamiento farmacológico , Glomérulos Renales/efectos de los fármacos , Microvasos/efectos de los fármacos , Animales , Ácidos Hidroxieicosatetraenoicos/toxicidad , Hipertensión/inducido químicamente , Hipertensión/patología , Glomérulos Renales/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Fenilefrina/toxicidad , Vasoconstrictores/toxicidad
18.
Pak J Pharm Sci ; 31(3): 747-754, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29716851

RESUMEN

Pioglitazone, peroxisome proliferator-activated receptor (PPAR-γ) agonist, is a therapeutic drug for diabetes. Present study investigated the interaction between PPAR-γ and alpha adrenoceptors in modulating vasopressor responses to Angiotensin II (Ang II) and adrenergic agonists, in diabetic & non-diabetic Spontaneously Hypertensive Rats (SHRs). Diabetes was induced with an i.p injection of streptozotocin (40 mg/kg) in two groups (STZ-CON, STZ-PIO), whereas two groups remained non diabetic (ND-CO, ND-PIO). One diabetic and non-diabetic group received Pioglitazone (10mg/kg) orally for 21 days. On day 28, the animals were anaesthetized with sodium pentobarbitone (60mg/kg) and prepared for measurement of systemic haemodynamics. Basal mean arterial pressure of STZ-CON was higher than ND-CON, whereas following pioglitazone treatment, MAP was lower compared to respective controls. MAP responses to i.v administration of NA, PE, ME and ANG II were significantly lower in diabetic SHRs: STZ-CON vs ND-CON (35%). Pioglitazone significantly decreased responses to NA, PE, ME and ANG II in ND-PIO versus ND-CON by 63%. Responses to NA and ANG II were significantly attenuated in STZ-PIO vs. ND-PIO (40%). PPAR-γ regulates systemic hemodynamic in diabetic model and cross-talk relationship exists between PPAR-γ and α1-adrenoceptors, ANG II in systemic vasculature of SHRs.


Asunto(s)
Agonistas Adrenérgicos/toxicidad , Angiotensina II/toxicidad , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Pioglitazona/uso terapéutico , Vasoconstrictores/toxicidad , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Hemodinámica/efectos de los fármacos , Hemodinámica/fisiología , Hipertensión/sangre , Hipertensión/inducido químicamente , Hipoglucemiantes/uso terapéutico , Ratas , Ratas Endogámicas SHR
19.
Stroke ; 48(9): 2557-2564, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28698257

RESUMEN

BACKGROUND AND PURPOSE: Hypertension is the major risk factor for stroke. Recent work unveiled that hypertension is associated with chronic neuroinflammation; microglia are the major players in neuroinflammation, and the activated microglia elevate sympathetic nerve activity and blood pressure. This study is to understand how brain homeostasis is kept from hypertensive disturbance and microglial activation at the onset of hypertension. METHODS: Hypertension was induced by subcutaneous delivery of angiotensin II, and blood pressure was monitored in conscious animals. Microglial activity was analyzed by flow cytometry and immunohistochemistry. Antibody, pharmacological chemical, and recombinant cytokine were administered to the brain through intracerebroventricular infusion. Microglial depletion was performed by intracerebroventricular delivering diphtheria toxin to CD11b-diphtheria toxin receptor mice. Gene expression profile in sympathetic controlling nucleus was analyzed by customized qRT-PCR array. RESULTS: Transforming growth factor-ß (TGF-ß) is constitutively expressed in the brains of normotensive mice. Removal of TGF-ß or blocking its signaling before hypertension induction accelerated hypertension progression, whereas supplementation of TGF-ß1 substantially suppressed neuroinflammation, kidney norepinephrine level, and blood pressure. By means of microglial depletion and adoptive transfer, we showed that the effects of TGF-ß on hypertension are mediated through microglia. In contrast to the activated microglia in established hypertension, the resting microglia are immunosuppressive and important in maintaining neural homeostasis at the onset of hypertension. Further, we profiled the signature molecules of neuroinflammation and neuroplasticity associated with hypertension and TGF-ß by qRT-PCR array. CONCLUSIONS: Our results identify that TGF-ß-modulated microglia are critical to keeping brain homeostasis responding to hypertensive disturbance.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Encéfalo/efectos de los fármacos , Hipertensión/inmunología , Microglía/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Traslado Adoptivo , Angiotensina II/toxicidad , Animales , Presión Sanguínea/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Antígeno CD11b , Toxina Diftérica , Citometría de Flujo , Factor de Crecimiento Similar a EGF de Unión a Heparina , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/fisiopatología , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Norepinefrina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sistema Nervioso Simpático , Transcriptoma , Factor de Crecimiento Transformador beta1/inmunología , Vasoconstrictores/toxicidad
20.
Basic Res Cardiol ; 112(3): 21, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28258299

RESUMEN

Vasopressors are widely used in resuscitation, ventricular failure, and sepsis, and often induce pulmonary hypertension with undefined mechanisms. We hypothesize that vasopressor-induced pulmonary hypertension is caused by increased pulmonary blood volume and tested this hypothesis in dogs under general anesthesia. In normal hearts (model 1), phenylephrine (2.5 µg/kg/min) transiently increased right but decreased left cardiac output, associated with increased pulmonary blood volume (63% ± 11.8, P = 0.007) and pressures in the left atrium, pulmonary capillary, and pulmonary artery. However, the trans-pulmonary gradient and pulmonary vascular resistance remained stable. These changes were absent after decreasing blood volume or during right cardiac dysfunction to reduce pulmonary blood volume (model 2). During double-ventricle bypass (model 3), phenylephrine (1, 2.5 and 10 µg/kg/min) only slightly induced pulmonary vasoconstriction. Vasopressin (1U and 2U) dose-dependently increased pulmonary artery pressure (52 ± 8.4 and 71 ± 10.3%), but did not cause pulmonary vasoconstriction in normally beating hearts (model 1). Pulmonary artery and left atrial pressures increased during left ventricle dysfunction (model 4), and further increased after phenylephrine injection by 31 ± 5.6 and 43 ± 7.5%, respectively. In conclusion, vasopressors increased blood volume in the lung with minimal pulmonary vasoconstriction. Thus, this pulmonary hypertension is similar to the hemodynamic pattern observed in left heart diseases and is passive, due to redistribution of blood from systemic to pulmonary circulation. Understanding the underlying mechanisms may improve clinical management of patients who are taking vasopressors, especially those with coexisting heart disease.


Asunto(s)
Hemodinámica/fisiología , Hipertensión Pulmonar/inducido químicamente , Circulación Pulmonar/fisiología , Vasoconstrictores/toxicidad , Animales , Perros
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda