Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Mol Cell Cardiol ; 121: 242-255, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30053525

RESUMEN

In hypertrophic hearts, autophagic flux insufficiency is recognized as a key pathology leading to maladaptive cardiac remodeling and heart failure. This study aimed to illuminate the cardioprotective role and mechanisms of a new myokine and adipokine, irisin, in cardiac hypertrophy and remodeling. Adult male wild-type, mouse-FNDC5 (irisin-precursor)-knockout and FNDC5 transgenic mice received 4 weeks of transverse aortic constriction (TAC) alone or combined with intraperitoneal injection of chloroquine diphosphate (CQ). Endogenous FNDC5 ablation aggravated and exogenous FNDC5 overexpression attenuated the TAC-induced hypertrophic damage in the heart, which was comparable to the protection of irisin against cardiomyocyte hypertrophy induced by angiotensin II (Ang II) or phenylephrine (PE). Accumulated autophagosome and impaired autophagy flux occurred in the TAC-treated myocardium and Ang II- or PE-insulted cardiomyocytes. Irisin deficiency caused reduced autophagy and aggravated autophagy flux failure, whereas irisin overexpression or supplementation induced protective autophagy and improved autophagy flux, which were reversed by autophagy inhibitors Atg5 siRNA, 3-MA and CQ. Irisin boosted the activity of only AMPK but not Akt and MAPK family members in hypertrophic hearts and cultured cardiomyocytes and further activated ULK1 at Ser555 but not Ser757 and did not affect the mTOR-S6K axis. Blockage of AMPK and ULK1 with compund C and SBI-0206965, respectively, both abrogated irisin's protection against cardiomyocyte hypertrophic injury and reversed its induction of both autophagy and autophagy flux. Our results suggest that irisin protects against pressure overload-induced cardiac hypertrophy by inducing protective autophagy and autophagy flux via activating AMPK-ULK1 signaling.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Cardiomegalia/genética , Fibronectinas/genética , Insuficiencia Cardíaca/genética , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Angiotensina II/administración & dosificación , Animales , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Benzamidas/administración & dosificación , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/patología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/efectos de los fármacos , Fenilefrina/administración & dosificación , Presión , Pirimidinas/administración & dosificación , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
2.
Basic Res Cardiol ; 111(2): 13, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26786260

RESUMEN

SIRT6, a member of the NAD(+)-dependent class III deacetylase sirtuin family, has been revealed to play important roles in promoting cellular resistance against oxidative stress. The formation of reactive oxygen species (ROS) and oxidative stress are the crucial mechanisms underlying cellular damage and dysfunction in cardiac ischemia/reperfusion (I/R) injury, but the role of SIRT6 in I/R-induced ROS and oxidative stress is poorly understood. In this study, by using heterozygous SIRT6 knockout (SIRT6(+/-)) mice and cultured neonatal cardiomyocyte models, we investigated how SIRT6 mediates oxidative stress and myocardial injury during I/R. Partial knockout (KO) of SIRT6 aggravated myocardial damage, ventricular remodeling, and oxidative stress in mice subjected to myocardial I/R, whereas restoration of SIRT6 expression by direct cardiac injection of adenoviral constructs encoding SIRT6 reversed these deleterious effects of SIRT6 KO in the ischemic heart. In addition, partial deletion of the SIRT6 gene decreased myocardial functional recovery following I/R in a Langendorff perfusion model. Similarly, the protective effects of SIRT6 were also observed in cultured cardiomyocytes following hypoxia/reoxygenation. Intriguingly, SIRT6 was noticed to up-regulate AMP/ATP and then activate the adenosine 5'-monophosphate-activated protein kinase (AMPK)-forkhead box O3α (FoxO3α) axis and further initiated the downstream antioxidant-encoding gene expression (manganese superoxide dismutase and catalase), thereby decreasing cellular levels of oxidative stress and mediating cardioprotection in the ischemic heart. These results suggest that SIRT6 protects the heart from I/R injury through FoxO3α activation in the ischemic heart in an AMP/ATP-induced AMPK-dependent way, thus upregulating antioxidants and suppressing oxidative stress.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Sirtuinas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Antioxidantes/metabolismo , Apoptosis , Catalasa/metabolismo , Células Cultivadas , Regulación hacia Abajo , Proteína Forkhead Box O3 , Técnicas In Vitro , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sirtuinas/genética , Superóxido Dismutasa/metabolismo , Remodelación Ventricular
3.
Phytomedicine ; 106: 154427, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36088791

RESUMEN

BACKGROUND: Liver dysfunction and liver failure are serious complications of sepsis, directly leading to septic progression and death. Now, there is no specific therapeutics available for sepsis-related liver dysfunction. Prim-O-glucosylcimifugin (POG), a chromone richest in the roots of Saposhnikovia divaricata (Turcz.) Schischk, is usually used to treat headache, rheumatoid arthritis and tetanus. While, the underlying mechanisms of POG against sepsis-induced liver damage and dysfunction are still not clear. PURPOSE: To study the anti-sepsis effect of POG, and its pharmacological mechanism to protect liver injury by weakening the function of macrophages in septic livers through inhibiting NOD-like receptor protein 3 (NLRP3) inflammasome pathway. METHOD: In vivo experiments, septic mouse model was induced by cecal ligation and puncture (CLP), and then the mortality was detected, liver inflammatory damages and plasma biomarkers of liver injury were evaluated by histopathological staining and biochemical assays, respectively. In vitro experiments, mouse primary peritoneal macrophages were treated with lipopolysaccharide (LPS) and ATP, and then the activated-inflammasomes, macrophage migration and polarization were detected by ASC immunofluorescence staining, transwell and flow cytometry assays, respectively. NLRP3 inflammasome components NLRP3, caspase-1, IL-1ß and IL-18 protein expressions were detected using western blot assays, and the contents of IL-1ß and IL-18 were measured by ELISA assays. RESULTS: POG treatment significantly decreased the mortality, liver inflammatory damages, hepatocyte apoptosis and plasma biomarkers of liver injury in CLP-challenged male WT mice, which were comparable to those in ibuprofen (a putative anti-inflammatory drug)-supplemented septic male WT mice and septic NLRP3 deficient-male mice. POG supplementation significantly suppressed NLRP3 inflammasome activation in septic liver tissues and cultured macrophages, by significantly reducing NLRP3, cleaved-caspase-1, IL-1ß and IL-18 levels, the activated-inflammasome ASC specks, and macrophage infiltration and migration, as well as M1-like polarization, but significantly increasing M2-like polarization. These findings were similar to the pharmacological effects of ibuprofen, NLRP3 deficiency, and a special NLRP3 inhibitor, MCC950. CONCLUSION: POG protected against sepsis by inhibiting NLRP3 inflammasome-mediated macrophage activation in septic liver and attenuating liver inflammatory injury, indicating that it may be a potential anti-sepsis drug candidate.


Asunto(s)
Inflamasomas , Sepsis , Adenosina Trifosfato , Animales , Caspasa 1/metabolismo , Cromonas , Ibuprofeno , Interleucina-18 , Lipopolisacáridos , Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas NLR , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Sepsis/metabolismo
4.
IEEE Trans Image Process ; 30: 6434-6445, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34232880

RESUMEN

The channel redundancy of convolutional neural networks (CNNs) results in the large consumption of memories and computational resources. In this work, we design a novel Slim Convolution (SlimConv) module to boost the performance of CNNs by reducing channel redundancies. Our SlimConv consists of three main steps: Reconstruct, Transform, and Fuse. It aims to reorganize and fuse the learned features more efficiently, such that the method can compress the model effectively. Our SlimConv is a plug-and-play architectural unit that can be used to replace convolutional layers in CNNs directly. We validate the effectiveness of SlimConv by conducting comprehensive experiments on various leading benchmarks, such as ImageNet, MS COCO2014, Pascal VOC2012 segmentation, and Pascal VOC2007 detection datasets. The experiments show that SlimConv-equipped models can achieve better performances consistently, less consumption of memory and computation resources than non-equipped counterparts. For example, the ResNet-101 fitted with SlimConv achieves 77.84% top-1 classification accuracy with 4.87 GFLOPs and 27.96M parameters on ImageNet, which shows almost 0.5% better performance with about 3 GFLOPs and 38% parameters reduced.

5.
Pflugers Arch ; 458(3): 481-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19151996

RESUMEN

Recent developments in the diversification of voltage-gated calcium channel function center on the rapidly emerging role of the posttranscriptional mechanism of alternative splicing. A number of diseases have been found to relate to the dysfunction of alternatively spliced exons arising from either genetic mutations or alterations in the splicing machinery. Mutations in some genes associated with congenital diseases have been detected to reside in alternatively spliced exons. As such, the severity of tissue-selective pathology of the disease will depend on the level of expression of the alternatively spliced exons in that tissue, as well as the extent in the change in channel properties. Importantly, alteration in channel properties is affected by the backbone array of the combinatorial alternatively spliced exons within the channel. In other words, the context by which mutations or alternatively spliced exons are expressed is a great influence on the alteration of channel properties and as such physiology and disease. We reviewed here recent comprehension of alternative splicing of voltage-gated calcium channels and how such structural and functional diversity of voltage-gated calcium channels will aid to clarify the pathophysiology of relevant diseases. Such understandings will further provide guidance for novel treatment.


Asunto(s)
Empalme Alternativo/genética , Canales de Calcio/fisiología , Predisposición Genética a la Enfermedad/genética , Biología Molecular/tendencias , Sitios de Empalme de ARN/genética , Humanos
6.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2379-2392, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31167124

RESUMEN

BACKGROUND: Abnormalities of the L-arginine-nitric oxide pathway induce hypertension. 5-Lipoxygenase (5-LO) is the key enzyme involved in synthesis of leukotrienes (LTs). However, whether nitricoxide synthase dysfunction induces hypertensive vascular remodeling by regulating 5-LO activity and its downstream inflammatory metabolites remains unknown. METHODS AND RESULTS: Six-week L-NAME treatment significantly induced hypertension and vascular remodeling in both wild-type (WT) and 5-LO-knockout (5-LO-KO) mice, and blood pressure in caudal and carotid arteries was lower in 5-LO-KO than WT mice with L-NAME exposure. On histology, L-NAME induced less media thickness, media-to-lumen ratio, and collagen deposition and fewer Ki-67-positive vascular smooth muscle cells (VSMCs) but more elastin expression in thoracic and mesenteric aortas of 5-LO-KO than L-NAME-treated WT mice. L-NAME significantly increased LT content, including LTB4 and cysteinyl LT (CysLTs), in plasma and neutrophil culture supernatants from WT mice. On immunohistochemistry, L-NAME promoted the colocalization of 5-LO and 5-LO-activating protein on the nuclear envelope of cultured neutrophils, which was accompanied by elevated LT content in culture supernatants. In addition, LTs significantly promoted BrdU incorporation, migration and phenotypic modulation in VSMCs. CONCLUSION: L-NAME may activate the 5-LO/LT pathway in immune cells, such as neutrophils, and promote the products of 5-LO metabolites, including LTB4 and CysLTs, which aggravate vascular remodeling in hypertension. 5-LO deficiency may protect against hypertension and vascular remodeling by reducing levels of 5-LO downstream inflammatory metabolites.


Asunto(s)
Araquidonato 5-Lipooxigenasa/genética , Hipertensión/prevención & control , Remodelación Vascular , Animales , Aorta/metabolismo , Aorta/patología , Araquidonato 5-Lipooxigenasa/deficiencia , Presión Sanguínea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/patología , Leucotrieno A4/sangre , Leucotrieno A4/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , NG-Nitroarginina Metil Éster/metabolismo , NG-Nitroarginina Metil Éster/toxicidad , Neutrófilos/inmunología , Neutrófilos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley , Remodelación Vascular/efectos de los fármacos
7.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 20(3): 207-10, 2003 Jun.
Artículo en Zh | MEDLINE | ID: mdl-12778445

RESUMEN

OBJECTIVE: To study the distribution of ScrF1 restriction polymorphism in intron 2 of the 3-hydroxy-3-methylglutaryl coenzyme A(HMG-CoA) reductase gene in Chinese Han population and the association of the polymorphism with coronary heart disease(CHD). METHODS: HMG-CoA reductase genotyping was performed using polymerase chain reaction-restriction fragment polymorphism. RESULTS: HMG-CoA reductase allelic frequencies of A, a were 0.519, 0.481; 0.440, 0.560 in CHD group and control group respectively. There was no significant difference in frequencies of allele and genotype in ScrF1 polymorphism between CHD group and control group(P>0.05). However, the levels of plasma very low density lipoprotein (VLDL) and TG in CHD patients with AA genotype were higher than those in CHD patients with other genotypes(P<0.05). The frequencies of A, a alleles at ScrF1 polymorphic site were significantly different from those reported in European Caucasians (0.44 vs 0.55, 0.56 vs 0.45, P<0.05). CONCLUSION: No direct association was found between the ScrF1 polymorphism and CHD, but there is a significant correlation between the AA genotype of the HMG-CoA reductase gene and the levels of plasma VLDL and TG in CHD group.


Asunto(s)
Pueblo Asiatico/genética , Hidroximetilglutaril-CoA Reductasas/genética , Metabolismo de los Lípidos/genética , Polimorfismo Genético , Adulto , Anciano , Anciano de 80 o más Años , Análisis Químico de la Sangre , VLDL-Colesterol/sangre , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hidroximetilglutaril-CoA-Reductasas NADP-Dependientes/genética , Masculino , Persona de Mediana Edad , Triglicéridos/sangre
8.
Br J Pharmacol ; 160(7): 1631-40, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20649567

RESUMEN

BACKGROUND AND PURPOSE: As a calcium channel blocker, diltiazem acts mainly on the voltage-gated calcium channels, Ca(v)1.2, for its beneficial effects in cardiovascular diseases such as hypertension, angina and/or supraventricular arrhythmias. However, the effects of diltiazem on different isoforms of Ca(v)1.2 channels expressed in heart and vascular smooth muscles remain to be investigated. Here, we characterized the effects of diltiazem on the splice variants of Ca(v)1.2 channels, predominant in cardiac and vascular smooth muscles. EXPERIMENTAL APPROACH: Cardiac and smooth muscle isoforms of Ca(v)1.2 channels were expressed in human embryonic kidney cells and their electrophysiological properties were characterized using whole-cell patch-clamp techniques. KEY RESULTS: Under closed-channel and use-dependent block (0.03 Hz), cardiac splice variant Ca(v)1.2CM was less sensitive to diltiazem than two major smooth muscle splice variants, Ca(v)1.2SM and Ca(v)1.2b. Ca(v)1.2CM has a more positive half-inactivation potential than the smooth muscle channels, and diltiazem shifted it less to negative potential. Additionally, the current decay was slower in Ca(v)1.2CM channels. When we modified alternatively spliced exons of cardiac Ca(v)1.2CM channels into smooth muscle exons, we found that all three loci contribute to the different diltiazem sensitivity between cardiac and smooth muscle splice isoforms. CONCLUSIONS AND IMPLICATIONS: Alternative splicing of Ca(v)1.2 channels modifies diltiazem sensitivity in the heart and blood vessels. Gating properties altered by diltiazem are different in the three channels.


Asunto(s)
Empalme Alternativo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Diltiazem/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocardio/metabolismo , Potenciales de Acción/efectos de los fármacos , Secuencia de Aminoácidos , Canales de Calcio Tipo L/genética , Línea Celular , Clonación Molecular , Humanos , Datos de Secuencia Molecular , Músculo Liso Vascular/metabolismo , Técnicas de Placa-Clamp
9.
Hypertens Res ; 32(10): 861-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19680258

RESUMEN

Intermedin (IMD), a new calcitonin/calcitonin gene-related peptide family peptide with vasodilatory and positive inotropic properties, has multiple functions in regulating cardiovascular homeostasis and is of particular interest in the pathophysiology of myocardial ischemia/reperfusion (MI/R). We created a mouse model of MI/R by ligating the cardiac left anterior descending artery to study the possible pathophysiological role of IMD and its receptor complexes in MI/R. Compared with the control, infarcted mice showed increased content, mRNA and protein expression of IMD in plasma and cardiac tissue. The mRNA expression of the receptor activity-modifying protein 3 (RAMP3) gene increased very early, and the calcitonin receptor-like receptor and RAMP2 mRNA levels increased later after reperfusion. However, the RAMP1 gene expression did not change. The tissue IMD content was positively correlated with the diastolic blood pressure and negatively correlated with pulse pressure. In addition, exogenous IMD treatment significantly ameliorated the MI/R injury by rescuing the pulse pressure, inhibiting neutrophil infiltration in the peri-infarction area, and decreasing the creatine kinase and lactate dehydrogenase activities in plasma. Our results indicated that IMD was upregulated in the ischemic myocardium and may induce important beneficial cytoprotection against cardiac ischemic injury.


Asunto(s)
Daño por Reperfusión Miocárdica/patología , Neuropéptidos/biosíntesis , Secuencia de Aminoácidos , Animales , Presión Sanguínea/efectos de los fármacos , Creatina Quinasa/sangre , Frecuencia Cardíaca/efectos de los fármacos , Inmunohistoquímica , L-Lactato Deshidrogenasa/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Neuropéptidos/síntesis química , Neuropéptidos/genética , Infiltración Neutrófila/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA