Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Theranostics ; 12(10): 4718-4733, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832088

RESUMEN

Excessive sympathetic activity and norepinephrine (NE) release play crucial roles in the pathogeneses of hypertension. Sympathetic fibers innervate adventitia rather than media of arteries. However, the roles of NE in adventitial fibroblasts (AFs) are unknown. This study investigated the roles of NE in regulating AFs-derived extracellular vesicles (EVs) release and vascular smooth muscle cells (VSMCs) proliferation in hypertension. Methods: AFs and VSMCs were prepared from aorta of Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). AFs were treated with NE (10 µM) for 24 h (every 6 h, 4 times), and cultured in exosomes-depleted medium for 48 h. EVs were isolated from AFs medium with ultracentrifugation for identification and transfer to VSMCs. Results: NE promoted AFs phenotypic transformation and proliferation, which were prevented by α-receptor antagonist phentolamine rather than ß-receptor antagonist propranolol. NE-treated AFs conditioned medium stimulated VSMCs proliferation, which was inhibited by either exosome inhibitor GW4869 or phentolamine. NE increased small EVs number, diameter and angiotensin converting enzyme (ACE) contents. The NE-induced EVs release was abolished by GW4869. The EVs from NE-treated AFs stimulated VSMCs proliferation, which was prevented by angiotensin II type 1 receptor antagonist losartan. The EVs from the ACE knockdown-treated AFs showed lower ACE contents, and lost their roles in stimulating VSMCs proliferation. Conclusion: NE promotes AFs-derived small EVs release and ACE transfer, and then causes VSMCs proliferation in hypertension. Intervention of AFs-derived EVs release may be potential therapeutics for excessive sympathetic activation-related vascular remodeling in hypertension.


Asunto(s)
Vesículas Extracelulares , Hipertensión , Adventicia/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Vesículas Extracelulares/metabolismo , Fibroblastos/metabolismo , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Norepinefrina/metabolismo , Norepinefrina/farmacología , Fentolamina/metabolismo , Fentolamina/farmacología , Ratas , Ratas Endogámicas WKY
2.
Redox Biol ; 48: 102204, 2021 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-34883403

RESUMEN

Superoxide and vascular smooth muscle cells (VSMCs) migration and proliferation play crucial roles in the vascular remodeling. Vascular remodeling contributes to the development and complications of hypertension. Rho family GTPase 3 (RND3 or RhoE), an atypical small Rho-GTPase, is known to be involved in cancer development and metastasis. However, the roles of RND3 in superoxide production and cardiovascular remodeling are unknown. Here, we uncovered the critical roles of RND3 in attenuating superoxide production, VSMCs migration and proliferation, and vascular remodeling in hypertension and its underline mechanisms. VSMCs were isolated and prepared from thoracic aorta of Male Wistar-Kyoto rat (WKY) and spontaneously hypertensive rat (SHR). RND3 mRNA and protein expressions in arteries and VSMCs were down-regulated in SHR. RND3 overexpression in VSMCs reduced NAD(P)H oxidase (NOX) activity, NOX1 and NOX2 expressions, mitochondria superoxide generation, and H2O2 production in SHR. Moreover, the RND3 overexpression inhibited VSMCs migration and proliferation in SHR, which were similar to the effects of NOX1 inhibitor ML171 plus NOX2 inhibitor GSK2795039. Rho-associated kinase 1 (ROCK1) and RhoA expressions and myosin phosphatase targeting protein 1 (MYPT1) phosphorylation in VSMCs were increased in SHR, which were prevented by RND3 overexpression. ROCK1 overexpression promoted NOX1 and NOX2 expressions, superoxide and H2O2 production, VSMCs migration and proliferation in both WKY and SHR, which were attenuated by RND3 overexpression. Adenoviral-mediated RND3 overexpression in SHR attenuated hypertension, vascular remodeling and oxidative stress. These results indicate that RND3 attenuates VSMCs migration and proliferation, hypertension and vascular remodeling in SHR via inhibiting ROCK1-NOX1/2 and mitochondria superoxide signaling.

3.
Acta Pharmacol Sin ; 42(11): 1798-1807, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33589794

RESUMEN

Proliferation of vascular smooth muscle cells (VSMCs) greatly contributes to vascular remodeling in hypertension. This study is to determine the roles and mechanisms of miR-135a-5p intervention in attenuating VSMC proliferation and vascular remodeling in spontaneously hypertensive rats (SHRs). MiR-135a-5p level was raised, while fibronectin type III domain-containing 5 (FNDC5) mRNA and protein expressions were reduced in VSMCs of SHRs compared with those of Wistar-Kyoto rats (WKYs). Enhanced VSMC proliferation in SHRs was inhibited by miR-135a-5p knockdown or miR-135a-5p inhibitor, but exacerbated by miR-135a-5p mimic. VSMCs of SHRs showed reduced myofilaments, increased or even damaged mitochondria, increased and dilated endoplasmic reticulum, which were attenuated by miR-135a-5p inhibitor. Dual-luciferase reporter assay shows that FNDC5 was a target gene of miR-135a-5p. Knockdown or inhibition of miR-135a-5p prevented the FNDC5 downregulation in VSMCs of SHRs, while miR-135a-5p mimic inhibited FNDC5 expressions in VSMCs of both WKYs and SHRs. FNDC5 knockdown had no significant effects on VSMC proliferation of WKYs, but aggravated VSMC proliferation of SHRs. Exogenous FNDC5 or FNDC5 overexpression attenuated VSMC proliferation of SHRs, and prevented miR-135a-5p mimic-induced enhancement of VSMC proliferation of SHR. MiR-135a-5p knockdown in SHRs attenuated hypertension, normalized FNDC5 expressions and inhibited vascular smooth muscle proliferation, and alleviated vascular remodeling. These results indicate that miR-135a-5p promotes while FNDC5 inhibits VSMC proliferation in SHRs. Silencing of miR-135a-5p attenuates VSMC proliferation and vascular remodeling in SHRs via disinhibition of FNDC5 transcription. Either inhibition of miR-135a-5p or upregulation of FNDC5 may be a therapeutically strategy in attenuating vascular remodeling and hypertension.


Asunto(s)
Hipertensión/metabolismo , MicroARNs/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Remodelación Vascular/fisiología , Animales , Proliferación Celular/fisiología , Células Cultivadas , Hipertensión/patología , Masculino , MicroARNs/antagonistas & inhibidores , Músculo Liso Vascular/ultraestructura , Miocitos del Músculo Liso/ultraestructura , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
4.
Sheng Li Xue Bao ; 72(6): 724-729, 2020 Dec 25.
Artículo en Chino | MEDLINE | ID: mdl-33349829

RESUMEN

The traditional medical experiment based on animal studies fails to reflect competency-oriented goal, and is not closely combined with clinical and scientific research, which does not meet the need for early clinical and scientific training. In order to cultivate the first-class medical talents, medical experimental teaching should conform to the trend of modern medical education, innovate teaching ideas and models, and update the hardware and software in time. Therefore, our teaching center adopts the triad medical experimental system which consists of "animal experiments, human functional experiments, and electronic standardized patient (ESP)-based virtual simulation experiments", and uses one system to integrate basic and clinical medicine, practice and virtual learning, teaching and scientific training. The system retains the core content of traditional animal experiments, and includes the most mature and widely used human physiological experiments to increase students' learning experience. With medical simulation experiment, it explains the specific physiological and pathophysiological processes of human body to improve students' cognitive and thinking ability. Here, we provide a systematic description on our triad medical experimental system, and discuss the experience to establish this novel system.


Asunto(s)
Aprendizaje , Estudiantes , Animales , Humanos
5.
Sheng Li Xue Bao ; 72(6): 730-736, 2020 Dec 25.
Artículo en Chino | MEDLINE | ID: mdl-33349830

RESUMEN

Informatization is an effective way to promote the reform and innovation of higher education and improve its quality. Virtual simulation teaching is indispensable in the educational informatization. Here, we describe the development and current situation of virtual simulation teaching, and introduce electronic standardized patient (ESP) based-virtual human body system powered by the real-time human physiological parameters. We also discuss how to build an ESP-based community in the teaching of human physiology, preclinical integrated case learning and other teaching projects. These ESP-based virtual simulation projects display the advantages of interdisciplinary fusion and the combination of basic and clinical knowledge, and open up the third type of functional experiments. Therefore, ESP-based virtual simulation teaching platform presumably becomes a considerable option for the first-class course construction in physiology.


Asunto(s)
Aprendizaje , Interfaz Usuario-Computador , Simulación por Computador , Electrónica , Humanos
6.
Neurosci Bull ; 36(5): 463-474, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31989424

RESUMEN

Chemical stimulation of the kidney increases sympathetic activity and blood pressure in rats. The hypothalamic paraventricular nucleus (PVN) is important in mediating the excitatory renal reflex (ERR). In this study, we examined the role of molecular signaling in the PVN in mediating the capsaicin-induced ERR and sympathetic activation. Bilateral PVN microinjections were performed in rats under anesthesia. The ERR was elicited by infusion of capsaicin into the cortico-medullary border of the right kidney. The reflex was evaluated as the capsaicin-induced changes in left renal sympathetic nerve activity and mean arterial pressure. Blockade of angiotensin type 1 receptors with losartan or inhibition of angiotensin-converting enzyme with captopril in the PVN abolished the capsaicin-induced ERR. Renal infusion of capsaicin significantly increased NAD(P)H oxidase activity and superoxide anion production in the PVN, which were prevented by ipsilateral renal denervation or microinjection of losartan into the PVN. Furthermore, either scavenging of superoxide anions or inhibition of NAD(P)H oxidase in the PVN abolished the capsaicin-induced ERR. We conclude that the ERR induced by renal infusion of capsaicin is mediated by angiotensin type 1 receptor-related NAD(P)H oxidase activation and superoxide anion production within the PVN.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Capsaicina/farmacología , Riñón/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Reflejo/efectos de los fármacos , Superóxidos/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Acetofenonas/farmacología , Acetilcisteína/farmacología , Alopurinol/farmacología , Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Presión Sanguínea/fisiología , Captopril/farmacología , Ditiocarba/farmacología , Riñón/inervación , Riñón/fisiología , Losartán/farmacología , Masculino , NADPH Oxidasas/antagonistas & inhibidores , Compuestos Onio/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1 , Reflejo/fisiología
7.
Neurosci Bull ; 36(2): 143-152, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31392556

RESUMEN

Sympathetic activation and the kidney play critical roles in hypertension and chronic heart failure. The role of the kidney in sympathetic activation is still not well known. In this study, we revealed an excitatory renal reflex (ERR) in rats induced by chemical stimulation of the kidney that regulated sympathetic activity and blood pressure. The ERR was induced by renal infusion of capsaicin, and evaluated by the changes in renal sympathetic outflow, blood pressure, and heart rate. Renal infusion of capsaicin dose-dependently increased the contralateral renal sympathetic nerve activity, mean arterial pressure, and heart rate. Capsaicin in the cortico-medullary border had greater effects than in the cortex or medulla. Intravenous infusion of capsaicin had no significant effects. The effects of renal infusion of capsaicin were abolished by ipsilateral renal denervation, but were not affected by bilateral sinoaortic denervation. Renal infusion of capsaicin increased the ipsilateral renal afferent activity. The ERR was also induced by renal infusion of bradykinin, adenosine, and angiotensin II, but not by ATP. Renal infusion of capsaicin increased c-Fos expression in the paraventricular nucleus (PVN) of hypothalamus. Lesion of neurons in the PVN with kainic acid abolished the capsaicin-induced ERR. These findings indicate that chemical stimulation of kidney causes an excitatory reflex, leading to sympathetic activation, pressor response, and accelerated heart rate. The PVN is an important central nucleus in the pathway of the ERR.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Riñón/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos , Angiotensina II , Animales , Presión Arterial/efectos de los fármacos , Bradiquinina , Capsaicina/farmacología , Frecuencia Cardíaca/fisiología , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Reflejo , Estimulación Química
8.
J Extracell Vesicles ; 9(1): 1698795, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31839907

RESUMEN

Proliferation of vascular smooth muscle cells (VSMCs) plays crucial roles in vascular remodelling and stiffening in hypertension. Vascular adventitial fibroblasts are a key regulator of vascular wall function and structure. This study is designed to investigate the roles of adventitial fibroblasts-derived extracellular vesicles (EVs) in VSMC proliferation and vascular remodelling in normotensive Wistar-Kyoto rat (WKY) and spontaneously hypertensive rat (SHR), an animal model of human essential hypertension. EVs were isolated from aortic adventitial fibroblasts of WKY (WKY-EVs) and SHR (SHR-EVs). Compared with WKY-EVs, miR155-5p content was reduced, while angiotensin-converting enzyme (ACE) content was increased in SHR-EVs. WKY-EVs inhibited VSMC proliferation of SHR, which was prevented by miR155-5p inhibitor. SHR-EVs promoted VSMC proliferation of both strains, which was enhanced by miR155-5p inhibitor, but abolished by captopril or losartan. Dual luciferase reporter assay showed that ACE was a target gene of miR155-5p. MiR155-5p mimic or overexpression inhibited VSMC proliferation and ACE upregulation of SHR. WKY-EVs reduced ACE mRNA and protein expressions while SHR-EVs only increased ACE protein level in VSMCs of both strains. However, the SHR-EVs-derived from the ACE knockdown-treated adventitial fibroblasts lost the roles in promoting VSMC proliferation and ACE upregulation. Systemic miR155-5p overexpression reduced vascular ACE, angiotensin II and proliferating cell nuclear antigen levels, and attenuated hypertension and vascular remodelling in SHR. Repetitive intravenous injection of SHR-EVs increased blood pressure and vascular ACE contents, and promoted vascular remodelling in both strains, while WKY-EVs reduced vascular ACE contents and attenuated hypertension and vascular remodelling in SHR. We concluded that WKY-EVs-mediated miR155-5p transfer attenuates VSMC proliferation and vascular remodelling in SHR via suppressing ACE expression, while SHR-EVs-mediated ACE transfer promotes VSMC proliferation and vascular remodelling.

9.
Vascul Pharmacol ; 121: 106579, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31319161

RESUMEN

Foam cell formation and monocytes adhesion are key events in pathogenesis of atherosclerosis. Vascular smooth muscle cells (VSMCs) are an important origin of foam cells besides macrophages. Fibronectin type III domain containing protein 5 (FNDC5) is a protein, which induces browning of fat and attenuates glucose/lipid metabolic derangements in obese mice. The present study was designed to determine the roles of FNDC5 in inhibiting foam cell formation and monocyte adhesion in VSMCs and its underlying mechanisms. Oxidized low-density lipoprotein (oxLDL) was used to induce foam cell formation and monocyte adhesion in human aortic VSMCs. Foam cell formation was evaluated by intracellular lipid droplets, cholesterol contents, and mRNA levels of acyl-coenzyme A: cholesterol acyltransferase 1 (ACAT-1) and ATP binding cassette transporter A-1 (ABCA-1). Monocyte adhesion was evaluated by the number of monocytes adhered to VSMCs and mRNA levels of monocyte chemotactic protein-1 (MCP-1) and vascular cell adhesion molecule-1 (VCAM-1). FNDC5 inhibited oxLDL-induced foam cell formation, monocyte adhesion, ABCA-1 mRNA downregulation, and ACAT-1, MCP-1 and VCAM-1 mRNA upregulation in VSMCs. It inhibited oxLDL-induced p65-NFκB nuclear translocation, NLRP3 upregulation, caspase-1 and IL-1ß production. Inhibition of NFκB with BMS-345541 or inhibition of NLRP3 inflammasome with MCC950 showed similar effects to FNDC5 in attenuating the oxLDL-induced foam cell formation, monocyte adhesion, and caspase-1 and IL-1ß production. The oxLDL-induced NLRP3 upregulation was prevented by BMS-345541 rather than MCC950. These results indicate that FNDC5 inhibits oxLDL-induced foam cell formation and monocyte adhesion in VSMCs via suppressing NFκB-mediated NLRP3 upregulation and IL-1ß production.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Fibronectinas/metabolismo , Células Espumosas/efectos de los fármacos , Lipoproteínas LDL/farmacología , Monocitos/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Fibronectinas/genética , Fibronectinas/farmacología , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Transducción de Señal , Células THP-1 , Regulación hacia Arriba
10.
Oxid Med Cell Longev ; 2019: 5018410, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30805081

RESUMEN

Proliferation and oxidative stress of vascular smooth muscle cells (VSMCs) contribute to vascular remodeling in hypertension and several major vascular diseases. B-cell lymphoma 6 (BCL6) functions as a transcriptional repressor. The present study is designed to determine the roles of BCL6 in VSMC proliferation and oxidative stress and underlying mechanism. Angiotensin (Ang) II was used to induce VSMC proliferation and oxidative stress in human VSMCs. Effects of BCL6 overexpression and knockdown were, respectively, investigated in Ang II-treated human VSMCs. Therapeutical effects of BCL6 overexpression on vascular remodeling, oxidative stress, and proliferation were determined in the aorta of spontaneously hypertensive rats (SHR). Ang II reduced BCL6 expression in human VSMCs. BCL6 overexpression attenuated while BCL6 knockdown enhanced the Ang II-induced upregulation of NADPH oxidase 4 (NOX4), production of reactive oxygen species (ROS), and proliferation of VSMCs. BCL6 expression was downregulated in SHR. BCL6 overexpression in SHR reduced NOX4 expression, ROS production, and proliferation of the aortic media of SHR. Moreover, BCL6 overexpression attenuated vascular remodeling and hypertension in SHR. However, BCL6 overexpression had no significant effects on NOX2 expression in human VSMCs or in SHR. We conclude that BCL6 attenuates proliferation and oxidative stress of VSMCs in hypertension.


Asunto(s)
Hipertensión/metabolismo , Hipertensión/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Angiotensina II/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/patología , Aorta/fisiopatología , Presión Sanguínea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Hipertensión/fisiopatología , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Remodelación Vascular/efectos de los fármacos
11.
Hypertension ; 72(4): 881-888, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354715

RESUMEN

Migration of vascular smooth muscle cells (VSMCs) is pivotal for vascular remodeling in hypertension. Vascular adventitial fibroblasts (AFs) are important in the homeostasis of vascular structure. This study is designed to investigate the roles of AF exosomes (AFE) in VSMC migration and underling mechanism. Primary VSMCs and AFs were obtained from the aorta of spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats. VSMC migration was evaluated with Boyden chamber assay and wound healing assay. AFE from WKY rats and SHR were isolated and identified. AFE from SHR promoted but AFE from WKY rats had no significant effect on VSMC migration. The effects of AFE on VSMC migration were prevented by an exosome inhibitor GW4869, an AT1R (Ang II [angiotensin II] type 1 receptor) antagonist losartan, or an inhibitor of ACE (angiotensin-converting enzyme) captopril. ACE contents and activity were much higher in AFE from SHR than those from WKY rats. There were no significant difference in Ang II and AT1R mRNA and protein levels between AFE from SHR and AFE from WKY rats. AFE from SHR increased Ang II and ACE contents and ACE activity in VSMCs of WKY rats and SHR. The changes of Ang II contents and ACE activity were prevented by captopril. ACE knockdown in AFs reduced ACE contents and activity in AFE from SHR and inhibited AFE-induced migration of VSMCs of WKY rats and those of SHR. These results indicate that exosomes from AFs of SHR transfer ACE to VSMCs, which increases Ang II levels and activates AT1R in VSMCs and thereby promotes VSMC migration.


Asunto(s)
Adventicia , Exosomas , Fibroblastos , Hipertensión , Peptidil-Dipeptidasa A , Remodelación Vascular , Adventicia/efectos de los fármacos , Adventicia/fisiología , Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Compuestos de Anilina/farmacología , Animales , Compuestos de Bencilideno/farmacología , Captopril/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Exosomas/efectos de los fármacos , Exosomas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Técnicas de Silenciamiento del Gen/métodos , Hipertensión/metabolismo , Hipertensión/patología , Hipertensión/fisiopatología , Losartán/farmacología , Músculo Liso Vascular/fisiología , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/fisiología
12.
Metabolism ; 83: 31-41, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29374559

RESUMEN

BACKGROUND: Obesity-induced chronic inflammation is critical in the pathogenesis of insulin resistance, and the recruitment and proinflammatory activation of adipose tissue macrophages (ATMs) is important for the development of this process. Here, we examined the effects of fibronectin type III domain-containing 5 (FNDC5) on inflammation and insulin resistance in high-fat diet-induced obese mice. MATERIALS AND METHODS: Male wild-type (WT) and FNDC5-/- mice were fed with standard chow (Ctrl) or high fat diet (HFD) for 20 weeks to induce obesity and insulin resistance. Firstly, effects of FNDC5 gene deletion on obesity, insulin resistance, macrophage accumulation and polarization and adipose tissue inflammation were determined in mice. Secondly, the macrophage polarity shift was further examined with flow cytometry in isolated stromal vascular fraction (SVF). Thirdly, the effects of exogenous FNDC5 on lipopolysaccharide (LPS)-induced macrophage polarization, inflammation and the underlying signaling mechanism were investigated in RAW264.7 macrophages and primary mouse peritoneal cavity macrophages (PMs). Finally, the therapeutic effects of FNDC5 overexpression were examined in HFD-induced obese WT and FNDC5-/- mice. RESULTS: FNDC5 gene deletion aggravated obesity, insulin resistance, fat accumulation and inflammation accompanied with enhanced AMPK inhibition, macrophages recruitment and M1 polarization in mice fed with HFD. Exogenous FNDC5 inhibited LPS-induced M1 macrophage polarization and inflammatory cytokine production via AMPK phosphorylation in both RAW264.7 macrophages and PMs. FNDC5 overexpression attenuated insulin resistance, AMPK inhibition, M1 macrophage polarization and inflammatory cytokine production in adipose tissue of obese WT and FNDC5-/- mice. CONCLUSIONS: FNDC5 attenuates adipose tissue inflammation and insulin resistance via AMPK-mediated macrophage polarization in HFD-induced obesity. FNDC5 plays several beneficial roles in obesity and may be used as a therapeutic regimen for preventing inflammation and insulin resistance in obesity and diabetes.


Asunto(s)
Tejido Adiposo/patología , Fibronectinas/fisiología , Inflamación/genética , Resistencia a la Insulina/genética , Activación de Macrófagos/genética , Obesidad , Adenilato Quinasa/fisiología , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Polaridad Celular/genética , Dieta Alta en Grasa , Fibronectinas/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/genética , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología
13.
Cell Death Dis ; 8(10): e3074, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28981106

RESUMEN

Inflammation is involved in pathogenesis of hypertension. NLRP3 inflammasome activation is a powerful mediator of inflammatory response via caspase-1 activation. The present study was designed to determine the roles and mechanisms of NLRP3 inflammasome in phenotypic modulation and proliferation of vascular smooth muscle cells (VSMCs) in hypertension. Experiments were conducted in spontaneously hypertensive rats (SHR) and primary aortic VSMCs. NLRP3 inflammasome activation was observed in the media of aorta in SHR and in the VSMCs from SHR. Knockdown of NLRP3 inhibited inflammasome activation, VSMC phenotypic transformation and proliferation in SHR-derived VSMCs. Increased NFκB activation, histone acetylation and histone acetyltransferase expression were observed in SHR-derived VSMCs and in media of aorta in SHR. Chromatin immunoprecipitation analysis revealed the increased histone acetylation, p65-NFκB and Pol II occupancy at the NLRP3 promoter in vivo and in vitro. Inhibition of NFκB with BAY11-7082 or inhibition of histone acetyltransferase with curcumin prevented the NLRP3 inflammasome activation, VSMC phenotype switching and proliferation in VSMCs from SHR. Moreover, curcumin repressed NFκB activation. Silencing of NLRP3 gene ameliorated hypertension, vascular remodeling, NLRP3 inflammasome activation and phenotype switching in the aorta of SHR. These results indicate that NLRP3 inflammasome activation response to histone acetylation and NFκB activation contributes to VSMC phenotype switching and proliferation and vascular remodeling in hypertension.


Asunto(s)
Proliferación Celular/genética , Hipertensión/genética , Inflamasomas/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Angiotensina II/genética , Animales , Caspasa 1/genética , Curcumina/administración & dosificación , Regulación de la Expresión Génica , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , FN-kappa B/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fenotipo , Ratas , Ratas Endogámicas SHR/genética , Ratas Endogámicas SHR/metabolismo , Transducción de Señal
14.
Diabetes ; 65(11): 3262-3275, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27504012

RESUMEN

Fibronectin type III domain-containing 5 (FNDC5) protein induces browning of subcutaneous fat and mediates the beneficial effects of exercise on metabolism. However, whether FNDC5 is associated with hepatic steatosis, autophagy, fatty acid oxidation (FAO), and lipogenesis remains unknown. Herein, we show the roles and mechanisms of FNDC5 in hepatic steatosis, autophagy, and lipid metabolism. Fasted FNDC5-/- mice exhibited severe steatosis, reduced autophagy, and FAO, and enhanced lipogenesis in the liver compared with wild-type mice. Energy deprivation-induced autophagy, FAO, and AMPK activity were attenuated in FNDC5-/- hepatocytes, which were restored by activating AMPK with 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR). Inhibition of mammalian target of rapamycin (mTOR) complex 1 with rapamycin enhanced autophagy and FAO and attenuated lipogenesis and steatosis in FNDC5-/- livers. FNDC5 deficiency exacerbated hyperlipemia, hepatic FAO and autophagy impairment, hepatic lipogenesis, and lipid accumulation in obese mice. Exogenous FNDC5 stimulated autophagy and FAO gene expression in hepatocytes and repaired the attenuated autophagy and palmitate-induced steatosis in FNDC5-/- hepatocytes. FNDC5 overexpression prevented hyperlipemia, hepatic FAO and autophagy impairment, hepatic lipogenesis, and lipid accumulation in obese mice. These results indicate that FNDC5 deficiency impairs autophagy and FAO and enhances lipogenesis via the AMPK/mTOR pathway. FNDC5 deficiency aggravates whereas FNDC5 overexpression prevents the HFD-induced hyperlipemia, hepatic lipid accumulation, and impaired FAO and autophagy in the liver.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína 5 Relacionada con la Autofagia/metabolismo , Fibronectinas/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Autofagia/genética , Autofagia/fisiología , Proteína 5 Relacionada con la Autofagia/genética , Western Blotting , Células Cultivadas , Fibronectinas/deficiencia , Fibronectinas/genética , Hepatocitos/metabolismo , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Oxidación-Reducción , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología
15.
Antioxid Redox Signal ; 24(18): 1045-57, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-26952533

RESUMEN

AIMS: Media-to-intima migration of vascular smooth muscle cells (VSMCs) is critical to intimal thickening in atherosclerosis and restenosis after coronary angioplasty. The aim of this study is to determine the effects of salusin-ß on VSMC migration and intimal hyperplasia after vascular injury and the underlying mechanism. RESULTS: In vitro, salusin-ß promoted VSMC migration, which was attenuated by matrix metalloproteinase (MMP)-9 inhibition. Inhibition or knockdown of p65-nuclear factor kappa beta (NFκB) in VSMCs suppressed salusin-ß-induced MMP-9 expression and VSMC migration. Salusin-ß increased NADPH oxidase 2 (NOX2) expression and reactive oxygen species (ROS) production, which were prevented by NOX2-small interfering RNA (siRNA) transfection. Salusin-ß-induced p65-NFκB translocation, MMP-9 expression, and VSMC migration were inhibited by ROS scavenger, NADPH oxidase inhibitor, or NOX2-siRNA. In vivo, carotid artery ligation-induced vascular injury resulted in intimal hyperplasia in injured artery in rats. Salusin-ß was upregulated in the injured carotid arteries of rats, which was attributed to reduced miR-133a-3p expression. Knockdown of salusin-ß with siRNA attenuated the vascular injury-induced intimal thickening, p65-NFκB nuclear translocation, and NOX2 and MMP-9 expressions in rats. INNOVATION: Salusin-ß is a critical modulator in VSMC migration and neointima formation in response to vascular injury. CONCLUSIONS: Salusin-ß promotes VSMC migration and vascular injury-induced intimal hyperplasia via MMP-9 accumulation due to NOX2 activation, followed by ROS production, IκBα phosphorylation and degradation, and p65-NFκB translocation. We propose that salusin-ß may be important in the VSMC migration and neointima of some vascular diseases. Antioxid. Redox Signal. 24, 1045-1057.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Metaloproteinasa 9 de la Matriz/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Arterias Carótidas/patología , Movimiento Celular , Células Cultivadas , Activación Enzimática , Inducción Enzimática , Masculino , Metaloproteinasa 9 de la Matriz/genética , Glicoproteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Transporte de Proteínas , Ratas Sprague-Dawley , Transducción de Señal , Túnica Íntima/patología , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
16.
Sci Rep ; 6: 21924, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26907958

RESUMEN

ß-aminoisobutyric acid (BAIBA) is a nature thymine catabolite, and contributes to exercise-induced protection from metabolic diseases. Here we show the therapeutical effects of BAIBA on hepatic endoplasmic reticulum (ER) stress and glucose/lipid metabolic disturbance in diabetes. Type 2 diabetes was induced by combined streptozotocin (STZ) and high-fat diet (HFD) in mice. Oral administration of BAIBA for 4 weeks reduced blood glucose and lipids levels, hepatic key enzymes of gluconeogenesis and lipogenesis expressions, attenuated hepatic insulin resistance and lipid accumulation, and improved insulin signaling in type 2 diabetic mice. BAIBA reduced hepatic ER stress and apoptosis in type 2 diabetic mice. Furthermore, BAIBA alleviated ER stress in human hepatocellular carcinoma (HepG2) cells with glucosamine-induced insulin resistance. Hepatic AMPK phosphorylation was reduced in STZ/HFD mice and glucosamine-treated HepG2 cells, which were restored by BAIBA treatment. The suppressive effects of BAIBA on glucosamine-induced ER stress were reversed by knockdown of AMPK with siRNA. In addition, BAIBA prevented thapsigargin- or tunicamycin-induced ER stress, and tunicamycin-induced apoptosis in HepG2 cells. These results indicate that BAIBA attenuates hepatic ER stress, apoptosis and glucose/lipid metabolic disturbance in mice with type 2 diabetes. AMPK signaling is involved to the role of BAIBA in attenuating ER stress.


Asunto(s)
Ácidos Aminoisobutíricos/farmacología , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Diabetes Mellitus Experimental/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Glucemia/análisis , Western Blotting , Colesterol/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa , Glucosamina/toxicidad , Células Hep G2 , Humanos , Inmunohistoquímica , Resistencia a la Insulina , Hígado/metabolismo , Hígado/patología , Ratones , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Tapsigargina/toxicidad , Triglicéridos/sangre , Tunicamicina/toxicidad
17.
Neuropharmacology ; 103: 247-56, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26746861

RESUMEN

Relaxin is recognized as an ovarian polypeptide hormone. Abundant relaxin binding sites are observed in hypothalamic paraventricular nucleus (PVN). This study was conducted to determine the roles and underlying mechanisms of relaxin in the PVN in sympathetic activation and hypertension in spontaneously hypertensive rats (SHR). Experiments were performed in normotensive Wistar-Kyoto rats (WKY) and SHR. Relaxin and its RXFP1 receptors in PVN were up-regulated in SHR. Relaxin-positive neurons existed in both parvocellular and magnocellular parts of the PVN. Presympathetic neurons and AVP neurons in the PVN expressed RXFP1, but not relaxin. Bilateral PVN microinjection of human relaxin-2 increased but anti-relaxin IgG reduced renal sympathetic nerve activity (RSNA), mean arterial pressure (MAP), plasma norepinephrine (NE) and arginine vasopressin (AVP) levels in SHR. The effects of relaxin-2 on RSNA and MAP were abolished by intravenous infusion of ganglionic blocker hexamethonium, and attenuated by AVP V1 receptor antagonist AAVP. Akt phosphorylation was enhanced in SHR, and relaxin-2 stimulated Akt phosphorylation and p85α subunit of PI3K expression. PI3K inhibitor LY294002 or Akt inhibitor MK-2206 abolished the effects of relaxin-2 on the RSNA, MAP and plasma NE, and attenuated the relaxin-2-induced AVP secretion. STAT5a and polymerase II (Pol II) binding to relaxin-promoter were significantly increased in SHR. Chronic PVN infusion of relaxin-2 with osmotic pumps in normal rats induced sympathetic activation, AVP secretion and hypertension accompanied with cardiovascular remodeling. Relaxin in the PVN contributes to sympathetic overdrive and hypertension via PI3K-Akt pathway.


Asunto(s)
Hipertensión/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Relaxina/metabolismo , Sistema Nervioso Simpático/metabolismo , Animales , Humanos , Hipertensión/inducido químicamente , Masculino , Neuronas/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Relaxina/administración & dosificación , Transducción de Señal , Sistema Nervioso Simpático/efectos de los fármacos
18.
Clin Sci (Lond) ; 129(10): 839-50, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26201094

RESUMEN

Increased glucose production and reduced hepatic glycogen storage contribute to metabolic abnormalities in diabetes. Irisin, a newly identified myokine, induces the browning of white adipose tissue, but its effects on gluconeogenesis and glycogenesis are unknown. In the present study, we investigated the effects and underlying mechanisms of irisin on gluconeogenesis and glycogenesis in hepatocytes with insulin resistance, and its therapeutic role in type 2 diabetic mice. Insulin resistance was induced by glucosamine (GlcN) or palmitate in human hepatocellular carcinoma (HepG2) cells and mouse primary hepatocytes. Type 2 diabetes was induced by streptozotocin/high-fat diet (STZ/HFD) in mice. In HepG2 cells, irisin ameliorated the GlcN-induced increases in glucose production, phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase) expression, and glycogen synthase (GS) phosphorylation; it prevented GlcN-induced decreases in glycogen content and the phosphoinositide 3-kinase (PI3K) p110α subunit level, and the phosphorylation of Akt/protein kinase B, forkhead box transcription factor O1 (FOXO1) and glycogen synthase kinase-3 (GSK3). These effects of irisin were abolished by the inhibition of PI3K or Akt. The effects of irisin were confirmed in mouse primary hepatocytes with GlcN-induced insulin resistance and in human HepG2 cells with palmitate-induced insulin resistance. In diabetic mice, persistent subcutaneous perfusion of irisin improved the insulin sensitivity, reduced fasting blood glucose, increased GSK3 and Akt phosphorylation, glycogen content and irisin level, and suppressed GS phosphorylation and PEPCK and G6Pase expression in the liver. Irisin improves glucose homoeostasis by reducing gluconeogenesis via PI3K/Akt/FOXO1-mediated PEPCK and G6Pase down-regulation and increasing glycogenesis via PI3K/Akt/GSK3-mediated GS activation. Irisin may be regarded as a novel therapeutic strategy for insulin resistance and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Fibronectinas/farmacología , Gluconeogénesis/efectos de los fármacos , Glucógeno/biosíntesis , Hepatocitos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Western Blotting , Células Cultivadas , Cromonas/farmacología , Fosfatidilinositol 3-Quinasa Clase I , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Fibronectinas/administración & dosificación , Fibronectinas/sangre , Gluconeogénesis/genética , Glucosa/metabolismo , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Glucógeno Sintasa/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Resistencia a la Insulina , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Morfolinas/farmacología , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
19.
Biochim Biophys Acta ; 1852(9): 1867-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26111885

RESUMEN

Irisin is a cleaved and secreted fragment of fibronectin type III domain containing 5 (FNDC5), and contributes to the beneficial effects of exercise on metabolism. Here we report the therapeutical effects of FNDC5/irisin on metabolic derangements and insulin resistance in obesity, and show the lipolysis effect of irisin and its signal molecular mechanism. In obese mice, lentivirus mediated-FNDC5 overexpression enhanced energy expenditure, lipolysis and insulin sensitivity, and reduced hyperlipidemia, hyperglycemia, hyperinsulinism, blood pressure and norepinephrine levels; it increased hormone-sensitive lipase (HSL) expression and phosphorylation, and reduced perilipin level and adipocyte diameter in adipose tissues. Subcutaneous perfusion of irisin reduced hyperlipidemia and hyperglycemia, and improved insulin resistance. Either FNDC5 overexpression or irisin perfusion only induced a tendency toward a slight decrease in body weight in obese mice. In 3T3-L1 adipocytes, irisin enhanced basal lipolysis rather than isoproterenol-induced lipolysis, which were prevented by inhibition of adenylate cyclase or PKA; irisin increased the HSL and perilipin phosphorylation; it increased PKA activity, and cAMP and HSL mRNA levels, but reduced perilipin expression. These results indicate that FNDC5/irisin ameliorates glucose/lipid metabolic derangements and insulin resistance in obese mice, and enhances lipolysis via cAMP-PKA-HSL/perilipin pathway. FNDC5 or irisin can be taken as an effective therapeutic strategy for metabolic disorders.

20.
Biochim Biophys Acta ; 1852(9): 1709-18, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26001930

RESUMEN

Vascular smooth muscle cell (VSMC) proliferation and vascular fibrosis are closely linked with hypertension and atherosclerosis. Salusin-ß is a bioactive peptide involved in the pathogenesis of atherosclerosis. However, it is still largely undefined whether salusin-ß is a potential candidate in the VSMC proliferation and vascular fibrosis. Experiments were carried out in human vascular smooth muscle cells (VSMCs) and in rats with intravenous injection of lentivirus expressing salusin-ß. In vitro, salusin-ß promoted VSMCs proliferation, which was attenuated by adenylate cyclase inhibitor SQ22536, PKA inhibitor Rp-cAMP, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor AG1478, ERK inhibitor U0126 or cAMP response element binding protein (CREB) inhibitor KG501. It promoted the phosphorylation of ERK1/2, CREB and EGFR, which were abolished by SQ22536 or Rp-cAMP. Furthermore, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor AG1478 diminished the salusin-ß-evoked ERK1/2 and CREB phosphorylation. On the other hand, salusin-ß increased collagen-I, collagen-III, fibronectin and connective tissue growth factor (CTGF) mRNA and phosphorylation of Smad2/3, which were prevented by ALK5 inhibitor A83-01. In vivo, salusin-ß overexpression increased the media thickness, media/lumen ratio coupled with ERK1/2, CREB, EGFR and Smad2/3 phosphorylation, as well as the mRNA of collagen-I, collagen-III, fibronectin, transforming growth factor-ß1 (TGF-ß1) and CTGF in arteries. Moreover, salusin-ß overexpression in rats caused severe hypertension. Intravenous injection of salusin-ß dose-relatedly increased blood pressure, but excessive salusin-ß decreased blood pressure and heart rate. These results indicate that salusin-ß promotes VSMC proliferation via cAMP-PKA-EGFR-CREB/ERK pathway and vascular fibrosis via TGF-ß1-Smad pathway. Increased salusin-ß contributes to vascular remodeling and hypertension.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...