Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Acta Pharmacol Sin ; 44(7): 1350-1365, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36737635

RESUMEN

Sympathetic stress is prevalent in cardiovascular diseases. Sympathetic overactivation under strong acute stresses triggers acute cardiovascular events including myocardial infarction (MI), sudden cardiac death, and stress cardiomyopathy. α1-ARs and ß-ARs, two dominant subtypes of adrenergic receptors in the heart, play a significant role in the physiological and pathologic regulation of these processes. However, little is known about the functional similarities and differences between α1- and ß-ARs activated temporal responses in stress-induced cardiac pathology. In this work, we systematically compared the cardiac temporal genome-wide profiles of acute α1-AR and ß-AR activation in the mice model by integrating transcriptome and proteome. We found that α1- and ß-AR activations induced sustained and transient inflammatory gene expression, respectively. Particularly, the overactivation of α1-AR but not ß-AR led to neutrophil infiltration at one day, which was closely associated with the up-regulation of chemokines, activation of NF-κB pathway, and sustained inflammatory response. Furthermore, there are more metabolic disorders under α1-AR overactivation compared with ß-AR overactivation. These findings provide a new therapeutic strategy that, besides using ß-blocker as soon as possible, blocking α1-AR within one day should also be considered in the treatment of acute stress-associated cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Receptores Adrenérgicos beta , Animales , Ratones , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Corazón , Arritmias Cardíacas , Inflamación/metabolismo , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo
2.
Nucleic Acids Res ; 49(5): 2522-2536, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33561291

RESUMEN

Simultaneous dysregulation of multiple microRNAs (miRs) affects various pathological pathways related to cardiac failure. In addition to being potential cardiac disease-specific markers, miR-23b/27b/24-1 were reported to be responsible for conferring cardiac pathophysiological processes. In this study, we identified a conserved guanine-rich RNA motif within the miR-23b/27b/24-1 cluster that can form an RNA G-quadruplex (rG4) in vitro and in cells. Disruption of this intragenic rG4 significantly increased the production of all three miRs. Conversely, a G4-binding ligand tetrandrine (TET) stabilized the rG4 and suppressed miRs production in human and rodent cardiomyocytes. Our further study showed that the rG4 prevented Drosha-DGCR8 binding and processing of the pri-miR, suppressing the biogenesis of all three miRs. Moreover, CRISPR/Cas9-mediated G4 deletion in the rat genome aberrantly elevated all three miRs in the heart in vivo, leading to cardiac contractile dysfunction. Importantly, loss of the G4 resulted in reduced targets for the aforementioned miRs critical for normal heart function and defects in the L-type Ca2+ channel-ryanodine receptor (LCC-RyR) coupling in cardiomyocytes. Our results reveal a novel mechanism for G4-dependent regulation of miR biogenesis, which is essential for maintaining normal heart function.


Asunto(s)
G-Cuádruplex , MicroARNs/química , MicroARNs/metabolismo , Contracción Miocárdica/genética , Miocitos Cardíacos/metabolismo , Animales , Bencilisoquinolinas/farmacología , Sistemas CRISPR-Cas , Células Cultivadas , G-Cuádruplex/efectos de los fármacos , Regulación de la Expresión Génica , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Sprague-Dawley , Ribonucleasa III/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
3.
Acta Pharmacol Sin ; 43(10): 2542-2549, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35354962

RESUMEN

Upon chronic stress, ß-adrenergic receptor activation induces cardiac fibrosis and leads to heart failure. The small molecule compound IMM-H007 has demonstrated protective effects in cardiovascular diseases via activation of AMP-activated protein kinase (AMPK). This study aimed to investigate IMM-H007 effects on cardiac fibrosis induced by ß-adrenergic receptor activation. Because adenosine analogs also exert AMPK-independent effects, we assessed AMPK-dependent and -independent IMM-H007 effects in murine models of cardiac fibrosis. Continual subcutaneous injection of isoprenaline for 7 days caused cardiac fibrosis and cardiac dysfunction in mice in vivo. IMM-H007 attenuated isoprenaline-induced cardiac fibrosis, diastolic dysfunction, α-smooth muscle actin expression, and collagen I deposition in both wild-type and AMPKα2-/- mice. Moreover, IMM-H007 inhibited transforming growth factor ß1 (TGFß1) expression in wild-type, but not AMPKα2-/- mice. By contrast, IMM-H007 inhibited Smad2/3 signaling downstream of TGFß1 in both wild-type and AMPKα2-/- mice. Surface plasmon resonance and molecular docking experiments showed that IMM-H007 directly interacts with TGFß1, inhibits its binding to TGFß type II receptors, and downregulates the Smad2/3 signaling pathway downstream of TGFß1. These findings suggest that IMM-H007 inhibits isoprenaline-induced cardiac fibrosis via both AMPKα2-dependent and -independent mechanisms. IMM-H007 may be useful as a novel TGFß1 antagonist.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Factor de Crecimiento Transformador beta1 , Proteínas Quinasas Activadas por AMP/metabolismo , Actinas/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Colágeno , Fibrosis , Isoproterenol/toxicidad , Ratones , Simulación del Acoplamiento Molecular , Receptores Adrenérgicos beta , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
4.
Acta Pharmacol Sin ; 43(3): 588-601, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33967278

RESUMEN

Cardiac hypertrophy is a common adaptive response to a variety of stimuli, but prolonged hypertrophy leads to heart failure. Hence, discovery of agents treating cardiac hypertrophy is urgently needed. In the present study, we investigated the effects of QF84139, a newly synthesized pyrazine derivative, on cardiac hypertrophy and the underlying mechanisms. In neonatal rat cardiomyocytes (NRCMs), pretreatment with QF84139 (1-10 µM) concentration-dependently inhibited phenylephrine-induced hypertrophic responses characterized by fetal genes reactivation, increased ANP protein level and enlarged cardiomyocytes. In adult male mice, administration of QF84139 (5-90 mg·kg-1·d-1, i.p., for 2 weeks) dose-dependently reversed transverse aortic constriction (TAC)-induced cardiac hypertrophy displayed by cardiomyocyte size, left ventricular mass, heart weights, and reactivation of fetal genes. We further revealed that QF84139 selectively activated the AMPK signaling pathway without affecting the phosphorylation of CaMKIIδ, ERK1/2, AKT, PKCε, and P38 kinases in phenylephrine-treated NRCMs and in the hearts of TAC-treated mice. In NRCMs, QF84139 did not show additive effects with metformin on the AMPK activation, whereas the anti-hypertrophic effect of QF84139 was abolished by an AMPK inhibitor Compound C or knockdown of AMPKα2. In AMPKα2-deficient mice, the anti-hypertrophic effect of QF84139 was also vanished. These results demonstrate that QF84139 attenuates the PE- and TAC-induced cardiac hypertrophy via activating the AMPK signaling. This structurally novel compound would be a promising lead compound for developing effective agents for the treatment of cardiac hypertrophy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Cardiomegalia/patología , Miocitos Cardíacos/efectos de los fármacos , Animales , Animales Recién Nacidos , Aorta/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Fenilefrina/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
Acta Pharmacol Sin ; 43(5): 1243-1250, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34349235

RESUMEN

ß-Adrenergic receptor (ß-AR) overactivation is a major pathological factor associated with cardiac diseases and mediates cardiac inflammatory injury. Glibenclamide has shown anti-inflammatory effects in previous research. However, it is unclear whether and how glibenclamide can alleviate cardiac inflammatory injury induced by ß-AR overactivation. In the present study, male C57BL/6J mice were treated with or without the ß-AR agonist isoprenaline (ISO) with or without glibenclamide pretreatment. The results indicated that glibenclamide alleviated ISO-induced macrophage infiltration in the heart, as determined by Mac-3 staining. Consistent with this finding, glibenclamide also inhibited ISO-induced chemokines and proinflammatory cytokines expression in the heart. Moreover, glibenclamide inhibited ISO-induced cardiac fibrosis and dysfunction in mice. To reveal the protective mechanism of glibenclamide, the NLRP3 inflammasome was further analysed. ISO activated the NLRP3 inflammasome in both cardiomyocytes and mouse hearts, but this effect was alleviated by glibenclamide pretreatment. Furthermore, in cardiomyocytes, ISO increased the efflux of potassium and the generation of ROS, which are recognized as activators of the NLRP3 inflammasome. The ISO-induced increases in these processes were inhibited by glibenclamide pretreatment. Moreover, glibenclamide inhibited the cAMP/PKA signalling pathway, which is downstream of ß-AR, by increasing phosphodiesterase activity in mouse hearts and cardiomyocytes. In conclusion, glibenclamide alleviates ß-AR overactivation-induced cardiac inflammation by inhibiting the NLRP3 inflammasome. The underlying mechanism involves glibenclamide-mediated suppression of potassium efflux and ROS generation by inhibiting the cAMP/PKA pathway.


Asunto(s)
Inflamasomas , Receptores Adrenérgicos beta , Animales , Arritmias Cardíacas , Gliburida/farmacología , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Potasio/metabolismo , Potasio/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores Adrenérgicos beta/metabolismo
6.
Acta Pharmacol Sin ; 41(3): 311-318, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31530901

RESUMEN

Acute sympathetic stress causes excessive secretion of catecholamines and induces cardiac injuries, which are mainly mediated by ß-adrenergic receptors (ß-ARs). However, α1-adrenergic receptors (α1-ARs) are also expressed in the heart and are activated upon acute sympathetic stress. In the present study, we investigated whether α1-AR activation induced cardiac inflammation and the underlying mechanisms. Male C57BL/6 mice were injected with a single dose of α1-AR agonist phenylephrine (PE, 5 or 10 mg/kg, s.c.) with or without pretreatment with α-AR antagonist prazosin (5 mg/kg, s.c.). PE injection caused cardiac dysfunction and cardiac inflammation, evidenced by the increased expression of inflammatory cytokine IL-6 and chemokines MCP-1 and MCP-5, as well as macrophage infiltration in myocardium. These effects were blocked by prazosin pretreatment. Furthermore, PE injection significantly increased the expression of NOD-like receptor protein 3 (NLRP3) and the cleavage of caspase-1 (p20) and interleukin-18 in the heart; similar results were observed in both Langendorff-perfused hearts and cultured cardiomyocytes following the treatment with PE (10 µM). Moreover, PE-induced NLRP3 inflammasome activation and cardiac inflammation was blocked in Nlrp3-/- mice compared with wild-type mice. In conclusion, α1-AR overactivation induces cardiac inflammation by activating NLRP3 inflammasomes.


Asunto(s)
Inflamasomas/metabolismo , Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Relación Dosis-Respuesta a Droga , Ecocardiografía , Corazón/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Fenilefrina/farmacología , Relación Estructura-Actividad , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/patología
7.
Eur Heart J ; 39(1): 60-69, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28549109

RESUMEN

Aims: Rapid over-activation of ß-adrenergic receptor (ß-AR) upon stress leads to cardiac inflammation, a prevailing factor that underlies heart injury. However, mechanisms by which acute ß-AR stimulation induce cardiac inflammation still remain unknown. Here, we set out to identify the crucial role of inflammasome/interleukin (IL)-18 in initiating and maintaining cardiac inflammatory cascades upon ß-AR insult. Methods and results: Male C57BL/6 mice were injected with a single dose of ß-AR agonist, isoproterenol (ISO, 5 mg/kg body weight) or saline subcutaneously. Cytokine array profiling demonstrated that chemokines dominated the initial cytokines upregulation specifically within the heart upon ß-AR insult, which promoted early macrophage infiltration. Further investigation revealed that the rapid inflammasome-dependent activation of IL-18, but not IL-1ß, was the critical up-stream regulator for elevated chemokine expression in the myocardium upon ISO induced ß1-AR-ROS signalling. Indeed, a positive correlation was observed between the serum levels of norepinephrine and IL-18 in patients with chest pain. Genetic deletion of IL-18 or the up-stream inflammasome component NLRP3 significantly attenuated ISO-induced chemokine expression and macrophage infiltration. In addition, IL-18 neutralizing antibodies selectively abated ISO-induced chemokines, proinflammatory cytokines and adhesion molecules but not growth factors. Moreover, blocking IL-18 early after ISO treatment effectively attenuated cardiac inflammation and fibrosis. Conclusion: Inflammasome-dependent activation of IL-18 within the myocardium upon acute ß-AR over-activation triggers cytokine cascades, macrophage infiltration and pathological cardiac remodelling. Blocking IL-18 at the early stage of ß-AR insult can successfully prevent inflammatory responses and cardiac injuries.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Inflamación/metabolismo , Interleucina-18/metabolismo , Miocardio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animales , Citocinas/metabolismo , Fibrosis/metabolismo , Corazón/efectos de los fármacos , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/inmunología , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/fisiología
8.
Sheng Li Xue Bao ; 71(2): 196-204, 2019 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-31008479

RESUMEN

Cell-to-cell connections provide conduits for signal exchanges, and play important functional roles in physiological and pathological processes of multicellular organisms. Membrane nanotubes are common long-distance connections between cells, not only transfer molecule signals and mitochondria, but also cooperate with gap junction and other cell-to-cell communications to transfer signals. During the last decade, there are many studies about membrane nanotubes, which focus on the similarities and differences between membrane nanotubes and other cell-to-cell communications, as well as their biological functions. In the present review, we summarized the latest findings about the structural diversity, the similarities and differences in signal transmission with other types of cell-to-cell communications, and physiological and pathological roles of membrane nanotubes.


Asunto(s)
Comunicación Celular , Membrana Celular/fisiología , Uniones Comunicantes/fisiología , Nanotubos , Humanos , Mitocondrias/fisiología
9.
Sheng Li Xue Bao ; 71(2): 225-234, 2019 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-31008482

RESUMEN

The autonomic nervous system consists of the sympathetic nervous system and the parasympathetic nervous system. These two systems control the heart and work in a reciprocal fashion to modulate myocardial energy metabolism, heart rate as well as blood pressure. Multiple cardiac pathological conditions are accompanied by autonomic imbalance, characterized by sympathetic overactivation and parasympathetic inhibition. Studies have shown that overactive sympathetic nervous system leads to increased cardiac inflammatory reaction. Orchestrated inflammatory response serves to clear dead cardiac tissue and activate reparative process, whereas excessive inflammation may result in pathological cardiac remodeling. Since the discovery of the α7 nicotinic acetylcholine receptor (α7nAChR)-mediated cholinergic anti-inflammatory pathway (CAP), the protective effects of the parasympathetic nervous system in cardiac inflammation have attracted more attention recently. In this review, we summarized the role and underlying mechanisms of the sympathetic and parasympathetic nervous systems in cardiac inflammation, in order to provide new insight into cardiac inflammatory response in cardiovascular diseases.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Corazón/fisiopatología , Inflamación/fisiopatología , Sistema Nervioso Parasimpático/fisiología , Humanos , Receptor Nicotínico de Acetilcolina alfa 7/fisiología
10.
Sheng Li Xue Bao ; 71(2): 248-260, 2019 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-31008484

RESUMEN

Prostaglandin (PG) E2 plays critical roles during pregnancy and parturition. Emerging evidence indicates that human labour is an inflammatory event. We sought to investigate the effect of PGE2 on the output of proinflammatory cytokines in cultured human uterine smooth muscle cells (HUSMCs) from term pregnant women and elucidate the role of subtypes of PGE2 receptors (EP1, EP2, EP3 and EP4). After drug treatment and/or transfection of each receptor siRNA, the concentrations of inflammatory secreting factors in HUSMCs culture medium were detected by the corresponding ELISA kits. The results showed that, PGE2 increased interleukin 6 (IL-6) and tumor necrosis factor alpha (TNFα) output, decreased chemokine (c-x-c motif) ligand 8 (CXCL8) output in a dose-dependent manner, but had no effect on IL-1ß and chemokine (c-c motif) ligand 2 (CCL-2) secretion of HUSMCs. EP1/EP3 agonist 17-phenyl-trinor-PGE2 stimulated IL-6 and TNFα whilst suppressing IL-1ß and CXCL8 output. The effects of 17-phenyl-trinor-PGE2 on IL-1ß and CXCL8 secretion were remained whereas its effect on IL-6 and TNFα output did not occur in the cells with EP3 knockdown. The stimulatory effects of 17-phenyl-trinor-PGE2 on IL-6 and TNFα were remained whereas the inhibitory effects of 17-phenyl-trinor-PGE2 on IL-1ß secretion was blocked in the cells with EP1 knockdown. Either of EP2 and EP4 agonists stimulated IL-1ß and TNFα output, which was reversed by EP2 and EP4 siRNA, respectively. The inhibitors of phospholipase C (PLC) and protein kinase C (PKC) blocked EP1/EP3 modulation of TNFα and CXCL8 output. PI3K inhibitor LY294002 and P38 inhibitor SB202190 blocked 17-phenyl-trinor-PGE2-induced IL-1ß and IL-6 output, respectively. The inhibitors of adenylyl cyclase and PKA prevented EP2 and EP4 stimulation of IL-1ß and TNFα output, whereas PLC and PKC inhibitors blocked EP2- and EP4-induced TNFα output but not IL-1ß output. Our data suggest that PGE2 receptors exhibit different effects on the output of various cytokines in myometrium, which can subtly modulate the inflammatory microenvironment in myometrium during pregnancy.


Asunto(s)
Citocinas/metabolismo , Miocitos del Músculo Liso/citología , Miometrio/citología , Receptores de Prostaglandina E/fisiología , Células Cultivadas , Cromonas/farmacología , Femenino , Humanos , Imidazoles/farmacología , Inflamación , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas , Embarazo , Piridinas/farmacología
11.
Sheng Li Xue Bao ; 70(1): 1-8, 2018 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-29492508

RESUMEN

MicroRNAs (miRNAs) are small noncoding RNAs that control diverse cellular and developmental events through repression of large sets of target mRNAs. miRNAs expressions were mainly regulated at two levels: transcriptional and post-transcriptional. Transcriptional regulation of miRNA-encoding genes produce specific expression patterns of individual miRNA. However, the mechanism of post-transcriptional regulation of miRNAs remains largely unknown. The present study was aimed to clarify whether HuR, an evolutionary conserved AU-rich binding protein, could regulate miRNAs expressions. By means of a computational screen for AUUUA motifs within pri-miRNAs, we found that the downstream of hsa-let-7c but not hsa-miR-21 was enriched of AUUUA motifs. Then we transfected HuR and mutant HuR lacking RNA recognition motif 3 (RRM3) respectively into HEK293T cells. And HuR protein and miRNAs expressions were detected by Western blot and real-time PCR, respectively. The results showed that the overexpression of HuR promoted mature hsa-let-7c expression but not hsa-miR-21 expression. Furthermore, overexpression of HuR deletion mutant lacking RRM3 did not promote hsa-let-7c expression. These results suggest that RRM3 is crucial for HuR mediating mature hsa-let-7c expression. Collectively, these findings proposed a novel role of HuR in biogenesis of miRNAs, possibly by way of post-transcriptional regulation of miRNAs.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato , Proteína 1 Similar a ELAV/genética , MicroARNs/genética , Motivo de Reconocimiento de ARN , Secuencia de Bases , Biología Computacional , Regulación de la Expresión Génica , Células HEK293 , Humanos
12.
Sheng Li Xue Bao ; 69(2): 123-128, 2017 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-28435970

RESUMEN

AMP-activated protein kinase (AMPK) activation has been shown to protect against fibrosis. However, the underlying mechanism remains unclear. Here we explored the effect of AMPK activation on transforming growth factor-ß1 (TGFß1) production induced by angiotensin II (AngII) in cardiac fibroblasts and the underlying mechanisms. Adult mouse cardiac fibroblasts were isolated. TGFß1 and AMPK activity were determined by ELISA and Western blots, respectively. Pretreatment of AMPK activator AICAR inhibited TGFß1 production induced by AngII in cardiac fibroblasts, which was reversed by AMPK inhibitor compound C. Furthermore, bioinformatics predicted a potential CCAAT/enhancer-binding protein ß (C/EBPß) binding site in the promoter region of the mouse Tgfb1 gene. Luciferase reporter with wild type, but not deleted, C/EBPß binding sites transfection in mouse embryonic fibroblasts showed increased TGFß1 transcriptional activity induced by AngII, indicating that C/EBPß mediates AngII-induced TGFß1 transcript expression. Pretreatment of AICAR inhibited C/EBPß expression induced by AngII. In conclusion, AMPK activation inhibited TGFß1 production induced by AngII in cardiac fibroblasts through targeting C/EBPß. This finding provides a new mechanism underlying the anti-fibrogenic effects of AMPK activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Fibroblastos/enzimología , Factor de Crecimiento Transformador beta1/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Angiotensina II/farmacología , Animales , Sitios de Unión , Células Cultivadas , Fibrosis , Corazón , Ratones , Miocardio/citología , Fosforilación , Ribonucleótidos/farmacología , Transfección
13.
J Biol Chem ; 289(13): 9449-59, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24515110

RESUMEN

It is well known that tumors damage affected tissues; however, the specific mechanism underlying such damage remains elusive. AMP-activated protein kinase (AMPK) senses energetic changes and regulates glucose metabolism. In this study, we examined the mechanisms by which AMPK promotes metabolic adaptation in the tumor-bearing liver using a murine model of colon cancer liver metastasis. Knock-out of AMPK α2 significantly enhanced tumor-induced glucose deprivation in the liver and increased the extent of liver injury and hepatocyte death. Mechanistically, we observed that AMPK α2 deficiency resulted in elevated reactive oxygen species, reduced mitophagy, and increased cell death in response to tumors or glucose deprivation in vitro. These results imply that AMPK α2 is essential for attenuation of liver injury during tumor metastasis via hepatic glucose deprivation and mitophagy-mediated inhibition of reactive oxygen species production. Therefore, AMPK α2 might represent an important therapeutic target for colon cancer metastasis-induced liver injury.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucosa/deficiencia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Hígado/lesiones , Hígado/metabolismo , Estrés Oxidativo , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Animales , Línea Celular Tumoral , Neoplasias del Colon/patología , Metabolismo Energético , Técnicas de Inactivación de Genes , Hepatocitos/citología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/patología , Neoplasias Hepáticas/patología , Ratones , Mitofagia , Necrosis/metabolismo , Especies Reactivas de Oxígeno/metabolismo
14.
J Biol Chem ; 289(14): 10084-94, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24554716

RESUMEN

Accumulating evidence suggests that growth differentiation factor 15 (GDF-15) is associated with the severity and prognosis of various cardiovascular diseases. However, the effect of GDF-15 on the regulation of cardiac remodeling is still poorly understood. In this present study, we demonstrate that GDF-15 blocks norepinephrine (NE)-induced myocardial hypertrophy through a novel pathway involving inhibition of EGFR transactivation. Both in vivo and in vitro assay indicate that NE was able to stimulate the synthesis of GDF-15. The up-regulation of GDF-15 feedback inhibits NE-induced myocardial hypertrophy, including quantitation of [(3)H]leucine incorporation, protein/DNA ratio, cell surface area, and ANP mRNA level. Further research shows that GDF-15 could inhibit the phosphorylation of EGF receptor and downstream kinases (AKT and ERK1/2) induced by NE. Clinical research also shows that serum GDF-15 levels in hypertensive patients were significant higher than in healthy volunteers and were positively correlated with the thickness of the posterior wall of the left ventricle, interventricular septum, and left ventricular mass, as well as the serum level of norepinephrine. In conclusion, NE induces myocardial hypertrophy and up-regulates GDF-15, and this up-regulation of GDF-15 negatively regulates NE-induced myocardial hypertrophy by inhibiting EGF receptor transactivation following NE stimulation.


Asunto(s)
Agonistas alfa-Adrenérgicos/efectos adversos , Cardiomegalia/sangre , Receptores ErbB/metabolismo , Factor 15 de Diferenciación de Crecimiento/sangre , Norepinefrina/efectos adversos , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Agonistas alfa-Adrenérgicos/administración & dosificación , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Femenino , Humanos , Masculino , Miocardio/metabolismo , Miocardio/patología , Norepinefrina/administración & dosificación , Estudios Retrospectivos
15.
Biochem Biophys Res Commun ; 462(2): 119-23, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25930996

RESUMEN

The AMP-activated protein kinase (AMPK) pathway is involved in regulating inflammation in several cell lines. We reported that fenoterol, a ß2-adrenergic receptor (ß2-AR) agonist, had anti-inflammatory effects in THP-1 cells, a monocytic cell line. Whether the fenoterol anti-inflammatory effect involves the AMPK pathway is unknown. In this study, we explored the mechanism of ß2-AR stimulation with fenoterol in a lipopolysaccharide (LPS)-induced inflammatory cytokine secretion in THP-1 cells. We studied whether fenoterol and ß-arrestin-2 or AMPKα1 subunit knockdown could affect LPS-induced AMPK activation, nuclear factor-kappa B (NF-κB) activation and inflammatory cytokine secretion. LPS-induced AMPK activation and interleukin 1ß (IL-1ß) release were reduced with fenoterol pretreatment of THP-1 cells. SiRNA knockdown of ß-arrestin-2 abolished the fenoterol inhibition of LPS-induced AMPK activation and interleukin 1ß (IL-1ß) release, thus ß-arrestin-2 mediated the anti-inflammatory effects of fenoterol on LPS-treated THP-1 cells. In addition, siRNA knockdown of AMPKα1 significantly attenuated the LPS-induced NF-κB activation and IL-1ß release, so AMPKα1 was a key signaling molecule involved in LPS-induced inflammatory cytokine production. These results suggested the ß2-AR agonist fenoterol inhibited LPS-induced AMPK activation and IL-1ß release via ß-arrestin-2 in THP-1 cells. The exploration of these mechanisms may help optimize therapeutic agents targeting these pathways in inflammatory diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Arrestinas/metabolismo , Citocinas/biosíntesis , Fenoterol/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Arrestinas/antagonistas & inhibidores , Arrestinas/genética , Línea Celular , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/biosíntesis , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Arrestina beta 2 , beta-Arrestinas
16.
Circ Res ; 112(4): 601-5, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23307820

RESUMEN

RATIONALE: During the transition from compensated hypertrophy to heart failure, the signaling between L-type Ca(2+) channels in the cell membrane/T-tubules and ryanodine receptors in the sarcoplasmic reticulum becomes defective, partially because of the decreased expression of a T-tubule-sarcoplasmic reticulum anchoring protein, junctophilin-2. MicroRNA (miR)-24, a junctophilin-2 suppressing miR, is upregulated in hypertrophied and failing cardiomyocytes. OBJECTIVE: To test whether miR-24 suppression can protect the structural and functional integrity of L-type Ca(2+) channel-ryanodine receptor signaling in hypertrophied cardiomyocytes. METHODS AND RESULTS: In vivo silencing of miR-24 by a specific antagomir in an aorta-constricted mouse model effectively prevented the degradation of heart contraction, but not ventricular hypertrophy. Electrophysiology and confocal imaging studies showed that antagomir treatment prevented the decreases in L-type Ca(2+) channel-ryanodine receptor signaling fidelity/efficiency and whole-cell Ca(2+) transients. Further studies showed that antagomir treatment stabilized junctophilin-2 expression and protected the ultrastructure of T-tubule-sarcoplasmic reticulum junctions from disruption. CONCLUSIONS: MiR-24 suppression prevented the transition from compensated hypertrophy to decompensated hypertrophy, providing a potential strategy for early treatment against heart failure.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Acoplamiento Excitación-Contracción/efectos de los fármacos , Insuficiencia Cardíaca/prevención & control , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , MicroARNs/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Oligonucleótidos Antisentido/uso terapéutico , Animales , Estenosis Aórtica Subvalvular/complicaciones , Canales de Calcio Tipo L/fisiología , Señalización del Calcio/fisiología , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/fisiología , Modelos Cardiovasculares , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Oligonucleótidos Antisentido/farmacología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/fisiología , Retículo Sarcoplasmático/ultraestructura
17.
BMC Gastroenterol ; 15: 32, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25887997

RESUMEN

BACKGROUND: The presence of diabetes mellitus (DM) is associated with increased liver morbidity and mortality risk in patients with chronic hepatitis B (CHB). Aim of this study was to identify factors associated with type 2 diabetes mellitus (T2DM) in CHB patients. METHODS: A cross-sectional study with pair-matched controls was conducted in Nantong Third People's Hospital, Nantong University, China. From January 2008 to December 2012, a total of 1783 CHB patients were screened for study subjects, among whom 207 patients with T2DM were enrolled as cases and 207 sex- and age-matched non-DM patients as controls. Demographic, anthropometric, lifestyle, clinical, and laboratory data were obtained from each subject. RESULTS: In the univariate model, thirteen variables showed marked differences between the DM group and non-DM group. Patients with longer duration of CHB (≥15 years) and alcoholic steatosis showed the highest likelihood of T2DM (odds ratio = 5.39 and 4.95; 95% confidence intervals 2.76-10.53 and 1.65-14.91). In the multivariate adjusted analysis, three CHB-related factors, namely high viral load, long duration of illness, and presence of cirrhosis, contributed to substantially increase the likelihood of T2DM, in addition to the other five risk factors including family history of DM, low education level, elevated triglycerides (TG), gamma-glutamyl transferase (GGT) levels, and presence of alcoholic steatosis. CONCLUSIONS: Our findings suggest that high viral load, long duration of CHB, presence of cirrhosis, alcoholic steatosis and several other factors may be potential risk factors for development of T2DM in CHB patients. It is of vital importance to monitor glucose in high-risk CHB patients and aggressively intervene on modifiable risk factors.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Hepatitis B Crónica/complicaciones , Adulto , Estudios de Casos y Controles , China/epidemiología , Estudios Transversales , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Escolaridad , Hígado Graso Alcohólico/epidemiología , Femenino , Hepatitis B Crónica/sangre , Hepatitis B Crónica/epidemiología , Humanos , Hipertrigliceridemia/epidemiología , Cirrosis Hepática/epidemiología , Masculino , Persona de Mediana Edad , Factores de Riesgo , Factores de Tiempo , Carga Viral , gamma-Glutamiltransferasa/sangre
18.
Am J Physiol Heart Circ Physiol ; 306(11): H1569-81, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24705558

RESUMEN

Inhibition of matrix metalloproteinases-2 (MMP-2) activation renders cardioprotection from ischemia/reperfusion (I/R) injury; however, the signaling pathways involved have not been fully understood. Intermittent hypobaric hypoxia (IHH) has been shown to enhance myocardial tolerance to I/R injury via triggering intrinsic adaptive responses. Here we investigated whether IHH protects the heart against I/R injury via the regulation of MMP-2 and how the MMP-2 is regulated. IHH (Po2 = 84 mmHg, 4-h/day, 4 wk) improved postischemic myocardial contractile performance, lactate dehydrogenase (LDH) release, and infarct size in isolated perfused rat hearts. Moreover, IHH reversed I/R-induced MMP-2 activation and release, disorders in the levels of MMP-2 regulators, peroxynitrite (ONOO(-)) and tissue inhibitor of metalloproteinase-4 (TIMP-4), and loss of the MMP-2 targets α-actinin and troponin I. This protection was mimicked, but not augmented, by a MMP inhibitor doxycycline and lost by the α1-adrenoceptor (AR) antagonist prazosin. Furthermore, IHH increased myocardial α1A-AR and α1B-AR density but not α1D-AR after I/R. Concomitantly, IHH further enhanced the translocation of PKC epsilon (PKCε) and decreased the release of mitochondrial cytochrome c due to I/R via the activation of α1B-AR but not α1A-AR or α1D-AR. IHH-conferred cardioprotection in the postischemic contractile function, LDH release, MMP-2 activation, and nitrotyrosine as well as TIMP-4 contents were mimicked but not additive by α1-AR stimulation with phenylephrine and were abolished by an α1B-AR antagonist chloroethylclonidine and a PKCε inhibitor PKCε V1-2. These findings demonstrate that IHH exerts cardioprotection through attenuating excess ONOO(-) biosynthesis and TIMP-4 loss and sequential MMP-2 activation via the activation of α1B-AR/PKCε pathway.


Asunto(s)
Hipoxia/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Daño por Reperfusión/metabolismo , Actinina/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Doxiciclina/farmacología , Masculino , Prazosina/farmacología , Proteína Quinasa C-epsilon/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Inhibidor Tisular de Metaloproteinasa-4
19.
Circ Res ; 111(7): 837-41, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22891046

RESUMEN

RATIONALE: Failing cardiomyocytes exhibit decreased efficiency of excitation-contraction (E-C) coupling. The downregulation of junctophilin-2 (JP2), a protein anchoring the sarcoplasmic reticulum to T-tubules, has been identified as a major mechanism underlying the defective E-C coupling. However, the regulatory mechanism of JP2 remains unknown. OBJECTIVE: To determine whether microRNAs regulate JP2 expression. METHODS AND RESULTS: Bioinformatic analysis predicted 2 potential binding sites of miR-24 in the 3'-untranslated regions of JP2 mRNA. Luciferase assays confirmed that miR-24 suppressed JP2 expression by binding to either of these sites. In the aortic stenosis model, miR-24 was upregulated in failing cardiomyocytes. Adenovirus-directed overexpression of miR-24 in cardiomyocytes decreased JP2 expression and reduced Ca(2+) transient amplitude and E-C coupling gain. CONCLUSIONS: MiR-24-mediated suppression of JP2 expression provides a novel molecular mechanism for E-C coupling regulation in heart cells and suggests a new target against heart failure.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Insuficiencia Cardíaca/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Regulación hacia Arriba , Animales , Estenosis de la Válvula Aórtica/patología , Calcio/metabolismo , Células Cultivadas , Biología Computacional , Acoplamiento Excitación-Contracción/fisiología , Insuficiencia Cardíaca/patología , Proteínas de la Membrana/genética , MicroARNs/genética , Modelos Animales , Miocitos Cardíacos/patología , ARN Mensajero/metabolismo , Ratas , Retículo Sarcoplasmático/fisiología
20.
Cardiovasc Drugs Ther ; 28(3): 221-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24852484

RESUMEN

PURPOSE: Relaxin, a new drug for heart failure therapy, exerts its cardiac actions through relaxin family peptide receptor 1 (RXFP1). Factors regulating RXFP1 expression remain unknown. We have investigated effects of activation of adrenoceptors (AR), an important modulator in the development and prognosis of heart failure, on expression of RXFP1 in rat cardiomyocytes and mouse left ventricles (LV). METHODS: Expression of RXFP1 at mRNA (real-time PCR) and protein levels (immunoblotting) was measured in cardiomyocytes treated with α- and ß-AR agonists or antagonists. RXFP1 expression was also determined in the LV of transgenic mouse strains with cardiac-restricted overexpression of α1A-, α1B- or ß2-AR. Specific inhibitors were used to explore signal pathways involved in α1-AR mediated regulation of RXFP1 in cardiomyocytes. RESULTS: In cultured cardiomyocytes, α1-AR stimulation resulted in 2-3 fold increase in RXFP1 mRNA (P < 0.001), which was blocked by specific inhibitors for protein kinase C (PKC) or mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK). Activation of ß1-, but not ß2-AR, significantly inhibited RXFP1 expression (P < 0.001). Relative to respective wild-type controls, RXFP1 mRNA levels in the LV of mice overexpressing α1A- or α1B-AR were increased by 3- or 10-fold, respectively, but unchanged in ß2-AR transgenic hearts. Upregulation by α1-AR stimulation RXFP1 expression was confirmed at protein levels both in vitro and in vivo. CONCLUSIONS: Expression of RXFP1 was up-regulated by α1-AR but suppressed by ß-AR, mainly ß1-AR subtype, in cardiomyocytes. Future studies are warranted to characterize the functional significance of such regulation, especially in the setting of heart failure.


Asunto(s)
Miocitos Cardíacos/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Péptidos/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Femenino , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos beta 1/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA