Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 970: 176510, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38493917

RESUMO

Activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway protects against N-methyl-D-aspartic acid (NMDA)-induced excitotoxic retinal injury. AMPK activation enhances fatty acid metabolism and ketone body synthesis. Ketone bodies are transported into neurons by monocarboxylate transporters (MCTs) and exert neuroprotective effects. In this study, we examined the distribution and expression levels of MCT1 and MCT2 in the retina and analyzed the effects of pharmacological inhibition of MCTs on the protective effects of metformin and 5-aminoimidazole-4-carboxamide (AICAR), activators of AMPK, against NMDA-induced retinal injury in rats. MCT1 was expressed in the blood vessels, processes of astrocytes and Müller cells, and inner segments of photoreceptors in the rat retina, whereas MCT2 was expressed in neuronal cells in the ganglion cell layer (GCL) and in astrocyte processes. The expression levels of MCT2, but not MCT1, decreased one day after intravitreal injection of NMDA (200 nmol). Intravitreal injection of NMDA decreased the number of cells in the GCL compared to the vehicle seven days after injection. Simultaneous injection of metformin (20 nmol) or AICAR (50 nmol) with NMDA attenuated NMDA-induced cell loss in the GCL, and these protective effects were attenuated by AR-C155858 (1 pmol), an inhibitor of MCTs. AR-C155858 alone had no significant effect on the retinal structure. These results suggest that AMPK-activating compounds protect against NMDA-induced excitotoxic retinal injury via mechanisms involving MCTs in rats. NMDA-induced neurotoxicity may be associated with retinal neurodegenerative changes in glaucoma and diabetic retinopathy. Therefore, AMPK-activating compounds may be effective in managing these retinal diseases.


Assuntos
Metformina , Doenças Retinianas , Tiofenos , Uracila/análogos & derivados , Ratos , Animais , Proteínas Quinases Ativadas por AMP/metabolismo , N-Metilaspartato/toxicidade , Ratos Sprague-Dawley , Retina/metabolismo , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/prevenção & controle , Doenças Retinianas/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Metformina/efeitos adversos
2.
J Cardiol ; 83(5): 323-329, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38266816

RESUMO

The heart utilizes glucose and its metabolites as both energy sources and building blocks for cardiac growth and survival under both physiological and pathophysiological conditions. YAP/TAZ, transcriptional co-activators of the Hippo pathway, are key regulators of cell proliferation, survival, and metabolism in many cell types. Increasing lines of evidence suggest that the Hippo-YAP/TAZ signaling pathway is involved in the regulation of both physiological and pathophysiological processes in the heart. In particular, YAP/TAZ play a critical role in mediating aerobic glycolysis, the Warburg effect, in cardiomyocytes. Here, we summarize what is currently known about YAP/TAZ signaling in the heart by focusing on the regulation of glucose metabolism and its functional significance.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Transativadores , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transativadores/metabolismo , Proteínas de Sinalização YAP , Glucose , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fosfoproteínas/metabolismo
3.
Neurosci Lett ; 793: 136999, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36470506

RESUMO

Resveratrol, a natural polyphenolic compound, reportedly possesses numerous biological activities, including anti-inflammatory and antioxidant effects. In the current study, we examined (1) the dilator effects of resveratrol on retinal arterioles, (2) the protective effects of resveratrol against excitotoxic retinal injury, and (3) whether these effects are mediated by the AMP-activated kinase (AMPK)-dependent pathway in rats. Male Wistar rats (7 to 10 weeks old) were used in this study. The diameters of the retinal arterioles, mean arterial pressure, and heart rate were measured in vivo. The retinal injury was assessed by histological examination. Intravenous injection of resveratrol (3 mg/kg) increased the diameter of the retinal arterioles without affecting the mean arterial pressure and heart rate. The AMPK inhibitor, compound C (5 mg/kg, intravenously), significantly attenuated the retinal vasodilator response to resveratrol. Seven days after intravitreal injection of N-methyl-d-aspartic acid (NMDA; 25, 50, and 100 nmol/eye), the number of cells located in the ganglion cell layer (GCL) was reduced, along with thinning of the inner plexiform layer. Intravitreal resveratrol injection (100 nmol/eye) reduced the NMDA (25 and 50 nmol/eye)-induced cell loss in the GCL. The neuroprotective effect of resveratrol was significantly but not completely reversed by compound C (10 nmol/eye). These results suggest that resveratrol dilates retinal arterioles and protects against NMDA-induced retinal neurodegeneration via an AMPK-dependent pathway in rats. Resveratrol may have the potential to slow the onset and progression of diseases associated with retinal ischemia by improving impaired retinal circulation and protecting retinal neuronal cells.


Assuntos
N-Metilaspartato , Resveratrol , Células Ganglionares da Retina , Animais , Masculino , Ratos , Proteínas Quinases Ativadas por AMP/metabolismo , Arteríolas/efeitos dos fármacos , N-Metilaspartato/efeitos adversos , N-Metilaspartato/farmacologia , Ratos Wistar , Resveratrol/farmacologia , Retina/metabolismo
4.
J Clin Invest ; 132(6)2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35133975

RESUMO

The heart utilizes multiple adaptive mechanisms to maintain pump function. Compensatory cardiac hypertrophy reduces wall stress and oxygen consumption, thereby protecting the heart against acute blood pressure elevation. The nuclear effector of the Hippo pathway, Yes-associated protein 1 (YAP), is activated and mediates compensatory cardiac hypertrophy in response to acute pressure overload (PO). In this study, YAP promoted glycolysis by upregulating glucose transporter 1 (GLUT1), which in turn caused accumulation of intermediates and metabolites of the glycolytic, auxiliary, and anaplerotic pathways during acute PO. Cardiac hypertrophy was inhibited and heart failure was exacerbated in mice with YAP haploinsufficiency in the presence of acute PO. However, normalization of GLUT1 rescued the detrimental phenotype. PO induced the accumulation of glycolytic metabolites, including l-serine, l-aspartate, and malate, in a YAP-dependent manner, thereby promoting cardiac hypertrophy. YAP upregulated the GLUT1 gene through interaction with TEA domain family member 1 (TEAD1) and HIF-1α in cardiomyocytes. Thus, YAP induces compensatory cardiac hypertrophy through activation of the Warburg effect.


Assuntos
Cardiomegalia , Miócitos Cardíacos , Proteínas de Sinalização YAP/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Ciclo do Ácido Cítrico , Transportador de Glucose Tipo 1/genética , Glicólise , Camundongos , Miócitos Cardíacos/metabolismo
5.
Circ Res ; 130(2): 234-248, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-34875852

RESUMO

BACKGROUND: During the development of heart failure, a fetal cardiac gene program is reactivated and accelerates pathological cardiac remodeling. We previously reported that a transcriptional repressor, NRSF (neuron restrictive silencer factor), suppresses the fetal cardiac gene program, thereby maintaining cardiac integrity. The underlying molecular mechanisms remain to be determined, however. METHODS: We aim to elucidate molecular mechanisms by which NRSF maintains normal cardiac function. We generated cardiac-specific NRSF knockout mice and analyzed cardiac gene expression profiles in those mice and mice cardiac-specifically expressing a dominant-negative NRSF mutant. RESULTS: We found that cardiac expression of Gαo, an inhibitory G protein encoded in humans by GNAO1, is transcriptionally regulated by NRSF and is increased in the ventricles of several mouse models of heart failure. Genetic knockdown of Gnao1 ameliorated the cardiac dysfunction and prolonged survival rates in these mouse heart failure models. Conversely, cardiac-specific overexpression of GNAO1 in mice was sufficient to induce cardiac dysfunction. Mechanistically, we observed that increasing Gαo expression increased surface sarcolemmal L-type Ca2+ channel activity, activated CaMKII (calcium/calmodulin-dependent kinase-II) signaling, and impaired Ca2+ handling in ventricular myocytes, which led to cardiac dysfunction. CONCLUSIONS: These findings shed light on a novel function of Gαo in the regulation of cardiac Ca2+ homeostasis and systolic function and suggest Gαo may be an effective therapeutic target for the treatment of heart failure.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Repressoras/genética
7.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R547-R562, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33112656

RESUMO

Mitochondria play key roles in the differentiation and maturation of human cardiomyocytes (CMs). As human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) hold potential in the treatment of heart diseases, we sought to identify key mitochondrial pathways and regulators, which may provide targets for improving cardiac differentiation and maturation. Proteomic analysis was performed on enriched mitochondrial protein extracts isolated from hiPSC-CMs differentiated from dermal fibroblasts (dFCM) and cardiac fibroblasts (cFCM) at time points between 12 and 115 days of differentiation, and from adult and neonatal mouse hearts. Mitochondrial proteins with a twofold change at time points up to 120 days relative to 12 days were subjected to ingenuity pathway analysis (IPA). The highest upregulation was in metabolic pathways for fatty acid oxidation (FAO), the tricarboxylic acid (TCA) cycle, oxidative phosphorylation (OXPHOS), and branched chain amino acid (BCAA) degradation. The top upstream regulators predicted to be activated were peroxisome proliferator-activated receptor γ coactivator 1 α (PGC1-α), the insulin receptor (IR), and the retinoblastoma protein (Rb1) transcriptional repressor. IPA and immunoblotting showed upregulation of the mitochondrial LonP1 protease-a regulator of mitochondrial proteostasis, energetics, and metabolism. LonP1 knockdown increased FAO in neonatal rat ventricular cardiomyocytes (nRVMs). Our results support the notion that LonP1 upregulation negatively regulates FAO in cardiomyocytes to calibrate the flux between glucose and fatty acid oxidation. We discuss potential mechanisms by which IR, Rb1, and LonP1 regulate the metabolic shift from glycolysis to OXPHOS and FAO. These newly identified factors and pathways may help in optimizing the maturation of iPSC-CMs.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/metabolismo , Biogênese de Organelas , Proteoma , Proteômica , Animais , Linhagem Celular , Linhagem da Célula , Metabolismo Energético , Humanos , Camundongos , Mitocôndrias Cardíacas/genética , Proteínas Mitocondriais/genética , Ratos , Fatores de Tempo
8.
JACC Basic Transl Sci ; 5(11): 1057-1069, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33294739

RESUMO

The treatment of pediatric heart failure is a long-standing unmet medical need. Angiotensin II supports mammalian perinatal circulation by activating cardiac L-type Ca2+ channels through angiotensin type 1 receptor (AT1R) and ß-arrestin. TRV027, a ß-arrestin-biased AT1R agonist, that has been reported to be safe but not effective for adult patients with heart failure, activates the AT1R/ß-arrestin pathway. We found that TRV027 evokes a long-acting positive inotropic effect specifically on immature cardiac myocytes through the AT1R/ß-arrestin/L-type Ca2+ channel pathway with minimum effect on heart rate, oxygen consumption, reactive oxygen species production, and aldosterone secretion. Thus, TRV027 could be utilized as a valuable drug specific for pediatric heart failure.

9.
Sci Rep ; 10(1): 11883, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32681032

RESUMO

Accumulating evidence suggests that human pluripotent stem cell-derived cardiomyocytes can affect "heart regeneration", replacing injured cardiac scar tissue with concomitant electrical integration. However, electrically coupled graft cardiomyocytes were found to innately induce transient post-transplant ventricular tachycardia in recent large animal model transplantation studies. We hypothesised that these phenomena were derived from alterations in the grafted cardiomyocyte characteristics. In vitro experiments showed that human embryonic stem cell-derived cardiomyocytes (hESC-CMs) contain nodal-like cardiomyocytes that spontaneously contract faster than working-type cardiomyocytes. When transplanted into athymic rat hearts, proliferative capacity was lower for nodal-like than working-type cardiomyocytes with grafted cardiomyocytes eventually comprising only relatively matured ventricular cardiomyocytes. RNA-sequencing of engrafted hESC-CMs confirmed the increased expression of matured ventricular cardiomyocyte-related genes, and simultaneous decreased expression of nodal cardiomyocyte-related genes. Temporal engraftment of electrical excitable nodal-like cardiomyocytes may thus explain the transient incidence of post-transplant ventricular tachycardia, although further large animal model studies will be required to control post-transplant arrhythmia.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Regeneração , Potenciais de Ação , Biomarcadores , Imunofluorescência , Expressão Gênica , Perfilação da Expressão Gênica , Imuno-Histoquímica , Filogenia
10.
Cardiovasc Res ; 116(10): 1742-1755, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31584633

RESUMO

AIMS: Thioredoxin 1 (Trx1) is an evolutionarily conserved oxidoreductase that cleaves disulphide bonds in oxidized substrate proteins such as mechanistic target of rapamycin (mTOR) and maintains nuclear-encoded mitochondrial gene expression. The cardioprotective effect of Trx1 has been demonstrated via cardiac-specific overexpression of Trx1 and dominant negative Trx1. However, the pathophysiological role of endogenous Trx1 has not been defined with a loss-of-function model. To address this, we have generated cardiac-specific Trx1 knockout (Trx1cKO) mice. METHODS AND RESULTS: Trx1cKO mice were viable but died with a median survival age of 25.5 days. They developed heart failure, evidenced by contractile dysfunction, hypertrophy, and increased fibrosis and apoptotic cell death. Multiple markers consistently indicated increased oxidative stress and RNA-sequencing revealed downregulation of genes involved in energy production in Trx1cKO mice. Mitochondrial morphological abnormality was evident in these mice. Although heterozygous Trx1cKO mice did not show any significant baseline phenotype, pressure-overload-induced cardiac dysfunction, and downregulation of metabolic genes were exacerbated in these mice. mTOR was more oxidized and phosphorylation of mTOR substrates such as S6K and 4EBP1 was impaired in Trx1cKO mice. In cultured cardiomyocytes, Trx1 knockdown inhibited mitochondrial respiration and metabolic gene promoter activity, suggesting that Trx1 maintains mitochondrial function in a cell autonomous manner. Importantly, mTOR-C1483F, an oxidation-resistant mutation, prevented Trx1 knockdown-induced mTOR oxidation and inhibition and attenuated suppression of metabolic gene promoter activity. CONCLUSION: Endogenous Trx1 is essential for maintaining cardiac function and metabolism, partly through mTOR regulation via Cys1483.


Assuntos
Metabolismo Energético , Insuficiência Cardíaca/metabolismo , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Tiorredoxinas/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Metabolismo Energético/genética , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/patologia , Miócitos Cardíacos/patologia , Estresse Oxidativo , Ratos Wistar , Transdução de Sinais , Tiorredoxinas/genética
11.
J Cardiovasc Pharmacol ; 74(6): 483-490, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31815864

RESUMO

The heart requires a high amount of energy, in the form of adenosine triphosphate, to maintain its viability and pump function. Anaerobic glycolysis and mitochondrial oxidative phosphorylation are the two main metabolic pathways by which adenosine triphosphate is generated, using fatty acids, glucose, lactate, and ketone bodies as primary substrates. Previous studies have demonstrated that, in response to stress, the heart undergoes alterations in metabolism, ranging from changes in substrate utilization to mitochondrial function, collectively called metabolic remodeling. However, the molecular mechanism mediating metabolic remodeling in the heart remains unclear. Yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), which are major downstream effectors of the Hippo signaling pathway, play an important role in the regulation of heart size and cellular homeostasis of cardiomyocytes through the regulation of various transcriptional factors under both physiological and pathophysiological conditions. Recent findings in various organs and cell types have revealed that YAP and TAZ play an important role in energy metabolism. Here, we summarize what is currently known about YAP/TAZ in the regulation of metabolism of various substrates and mitochondrial function in various organs and cell types and discuss the potential role of YAP/TAZ in mediating metabolic remodeling of the heart during stress and heart failure.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Metabolismo Energético , Insuficiência Cardíaca/metabolismo , Mitocôndrias Cardíacas/metabolismo , Miocárdio/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Adaptação Fisiológica , Animais , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Via de Sinalização Hippo , Humanos , Mitocôndrias Cardíacas/patologia , Miocárdio/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
13.
J Pharmacol Exp Ther ; 368(1): 50-58, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30409832

RESUMO

An injury of the somatosensory system causes neuropathic pain, which is usually refractory to conventional analgesics, thus warranting the development of novel drugs against this kind of pain. The mechanism of neuropathic pain in rats that had undergone left L5 spinal nerve transection was analyzed. Ten days after surgery, these rats acquired neuropathic pain. The patch-clamp technique was used on the isolated bilateral L5 dorsal root ganglion neurons. The current-clamped neurons on the ipsilateral side exhibited significantly higher excitability than those on the contralateral side. However, only neurons with diameters of 40-50 µm on the ipsilateral side exhibited significantly larger voltage sags in response to hyperpolarizing current pulses than those on the contralateral side. Under the voltage clamp, only these neurons on the ipsilateral side showed a significantly larger density of an inward current at < -80 mV [hyperpolarization-activated nonselective cation (I h) current] with a rightward-shifted activation curve than that on the contralateral side. Ivabradine-an I h current inhibitor-inhibited I h currents in these neurons on both sides in a similar concentration-dependent manner, with an IC50 value of ∼3 µM. Moreover, the oral administration of ivabradine significantly alleviated the neuropathic pain on the ipsilateral side. An inhibitor of adenylyl cyclase or an antagonist of prostanoid EP4 receptors (CJ-023423) inhibited ipsilateral, but not contralateral I h, currents in these neurons. Furthermore, the intrathecal administration of CJ-023423 significantly attenuated neuropathic pain on the ipsilateral side. Thus, ivabradine and/or CJ-023423 may be a lead compound for the development of novel therapeutics against neuropathic pain.


Assuntos
Gânglios Espinais/fisiologia , Neuralgia/fisiopatologia , Neurônios/fisiologia , Receptores de Prostaglandina E Subtipo EP4/fisiologia , Animais , Relação Dose-Resposta a Droga , Gânglios Espinais/efeitos dos fármacos , Injeções Espinhais , Ivabradina/administração & dosagem , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neuralgia/tratamento farmacológico , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Sulfonamidas/administração & dosagem
14.
Glycoconj J ; 36(1): 1-11, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30536036

RESUMO

Serum sulfatides are critical glycosphingolipids that are present in lipoproteins and exert anticoagulant effects. A previous study reported decreased levels of serum sulfatides in hemodialysis patients and suggested an association with cardiovascular disease. However, the mechanism of changes in serum sulfatides in chronic kidney dysfunction has not been well investigated. The current study examined whether a chronic kidney disease (CKD) state could decrease serum sulfatide levels using 5/6 nephrectomy (5/6NCKD) mice, an established CKD murine model, and studied the mechanisms contributing to diminished sulfatides. 5/6NCKD mice and sham operation control mice were sacrificed at the 4th or 12th postoperative week (POW) for measurement of serum sulfatide levels. Hepatic sulfatide content, which is the origin of serum sulfatides, and the expression of sulfatide metabolic enzymes in liver tissue were assessed as well. The 5/6NCKD mice developed CKD and showed increased serum creatinine and indoxyl sulfate. The serum levels and hepatic amounts of sulfatides were significantly decreased in 5/6NCKD mice at both 4 and 12 POW, while the degradative enzymes of sulfatides arylsulfatase A and galactosylceramidase were significantly increased. In a Hepa1-6 murine liver cell line, indoxyl sulfate addition caused intracellular levels of sulfatides to decrease and degradative enzymes of sulfatides to increase in a manner comparable to the changes in 5/6NCKD mice liver tissue. In conclusion, chronic kidney dysfunction causes degradation of sulfatides in the liver to decrease serum sulfatide levels. One explanation of these results is that indoxyl sulfate, a uremic toxin, accelerates the degradation of sulfatides in liver tissue.


Assuntos
Insuficiência Renal Crônica/sangue , Sulfoglicoesfingolipídeos/sangue , Animais , Linhagem Celular Tumoral , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência Renal Crônica/metabolismo , Sulfoglicoesfingolipídeos/metabolismo
15.
Hypertens Res ; 42(5): 598-609, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30531843

RESUMO

Serum sulfatides are critical glycosphingolipids present in lipoproteins that work as modulators of thrombosis and hemostasis. Decreased serum sulfatide levels are suggested by our previous work to be related to cardiovascular disease (CVD). Hypertension, known to be an important risk factor for CVD, may affect serum sulfatide levels. However, how hypertension affects serum sulfatides directly and mechanistically is unknown. To elucidate these possible mechanisms, we investigated changes in serum sulfatide levels and their metabolism using an established experimental model of hypertension that uses continuous infusion of angiotensin II (AngII) into mice. Furthermore, we also examined the effects of four different antihypertensive drugs (losartan, irbesartan, nifedipine, and hydralazine) on serum sulfatide metabolism. Serum levels of sulfatides were found to be decreased in groups in which only hypertension was induced (AngII only), whereas they were increased in groups with reduced blood pressure (antihypertensives only) and ameliorated to increasingly normal levels in groups with induced hypertension that were also treated (AngII+antihypertensives). Changes in serum sulfatides were strongly related to hepatic expression levels of cerebroside sulfotransferase (CST), which is a key enzyme involved in sulfatide synthesis. Furthermore, the current study suggests that the primary factors affecting CST expression are oxidative stress, peroxisome proliferator-activated receptor α activity and blood pressure itself. This study demonstrates that hypertension significantly decreases levels of serum sulfatides by reducing hepatic CST expression via various effects mediated by AngII. Antihypertensive treatments can ameliorate abnormalities in serum sulfatide levels and may partially prevent hypertension related CVD by positively affecting sulfatide metabolism.


Assuntos
Anti-Hipertensivos/farmacologia , Hipertensão/sangue , Sulfoglicoesfingolipídeos/sangue , Angiotensina II , Antagonistas de Receptores de Angiotensina , Animais , Anti-Hipertensivos/uso terapêutico , Hipertensão/tratamento farmacológico , Fígado/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/metabolismo , Sulfotransferases/metabolismo
16.
Proc Natl Acad Sci U S A ; 115(17): 4507-4512, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29632175

RESUMO

Close physical association of CaV1.1 L-type calcium channels (LTCCs) at the sarcolemmal junctional membrane (JM) with ryanodine receptors (RyRs) of the sarcoplasmic reticulum (SR) is crucial for excitation-contraction coupling (ECC) in skeletal muscle. However, the molecular mechanism underlying the JM targeting of LTCCs is unexplored. Junctophilin 1 (JP1) and JP2 stabilize the JM by bridging the sarcolemmal and SR membranes. Here, we examined the roles of JPs in localization and function of LTCCs. Knockdown of JP1 or JP2 in cultured myotubes inhibited LTCC clustering at the JM and suppressed evoked Ca2+ transients without disrupting JM structure. Coimmunoprecipitation and GST pull-down assays demonstrated that JPs physically interacted with 12-aa residues in the proximal C terminus of the CaV1.1. A JP1 mutant lacking the C terminus including the transmembrane domain (JP1ΔCT) interacted with the sarcolemmal/T-tubule membrane but not the SR membrane. Expression of this mutant in adult mouse muscles in vivo exerted a dominant-negative effect on endogenous JPs, impairing LTCC-RyR coupling at triads without disrupting JM morphology, and substantially reducing Ca2+ transients without affecting SR Ca2+ content. Moreover, the contractile force of the JP1ΔCT-expressed muscle was dramatically reduced compared with the control. Taken together, JPs recruit LTCCs to the JM through physical interaction and ensure robust ECC at triads in skeletal muscle.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/fisiologia , Proteínas de Membrana/metabolismo , Contração Muscular/fisiologia , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Linhagem Celular , Proteínas de Membrana/genética , Camundongos , Proteínas Musculares/genética , Domínios Proteicos , Sarcolema/genética , Sarcolema/metabolismo
17.
Pflugers Arch ; 470(6): 909-921, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29441404

RESUMO

In atherosclerosis, vascular smooth muscle cells (VSMC) migrate from the media toward the intima of the arteries in response to cytokines, such as platelet-derived growth factor (PDGF). However, molecular mechanism underlying the PDGF-induced migration of VSMCs remains unclear. The migration of rat aorta-derived synthetic VSMCs, A7r5, in response to PDGF was potently inhibited by a CaV1.2 channel inhibitor, nifedipine, and a Src family tyrosine kinase (SFK)/Abl inhibitor, bosutinib, in a less-than-additive manner. PDGF significantly increased CaV1.2 channel currents without altering CaV1.2 protein expression levels in A7r5 cells. This reaction was inhibited by C-terminal Src kinase, a selective inhibitor of SFKs. In contractile VSMCs, the C-terminus of CaV1.2 is proteolytically cleaved into proximal and distal C-termini (PCT and DCT, respectively). Clipped DCT is noncovalently reassociated with PCT to autoinhibit the channel activity. Conversely, in synthetic A7r5 cells, full-length CaV1.2 (CaV1.2FL) is expressed much more abundantly than truncated CaV1.2. In a heterologous expression system, c-Src activated CaV1.2 channels composed of CaV1.2FL but not truncated CaV1.2 (CaV1.2Δ1763) or CaV1.2Δ1763 plus clipped DCT. Further, c-Src enhanced the coupling efficiency between the voltage-sensing domain and activation gate of CaV1.2FL channels by phosphorylating Tyr1709 and Tyr1758 in PCT. Compared with CaV1.2Δ1763, c-Src could more efficiently bind to and phosphorylate CaV1.2FL irrespective of the presence or absence of clipped DCT. Therefore, in atherosclerotic lesions, phenotypic switching of VSMCs may facilitate pro-migratory effects of PDGF on VSMCs by suppressing posttranslational CaV1.2 modifications.


Assuntos
Aterosclerose/metabolismo , Canais de Cálcio Tipo L/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Migração Transendotelial e Transepitelial , Animais , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Canais de Cálcio Tipo L/química , Células Cultivadas , Células HEK293 , Humanos , Masculino , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Domínios Proteicos , Ratos , Ratos Sprague-Dawley , Quinases da Família src/metabolismo
18.
J Physiol ; 595(13): 4207-4225, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28295363

RESUMO

KEY POINTS: Angiotensin II (AngII) is crucial in cardiovascular regulation in perinatal mammalians. Here we show that AngII increases twitch Ca2+ transients of mouse immature but not mature cardiomyocytes by robustly activating CaV 1.2 L-type Ca2+ channels through a novel signalling pathway involving angiotensin type 1 (AT1 ) receptors, ß-arrestin2 and casein kinase 2. A ß-arrestin-biased AT1 receptor agonist, TRV027, was as effective as AngII in activating L-type Ca2+ channels. Our results help understand the molecular mechanism by which AngII regulates the perinatal circulation and also suggest that ß-arrestin-biased AT1 receptor agonists may be valuable therapeutics for paediatric heart failure. ABSTRACT: Angiotensin II (AngII), the main effector peptide of the renin-angiotensin system, plays important roles in cardiovascular regulation in the perinatal period. Despite the well-known stimulatory effect of AngII on vascular contraction, little is known about regulation of contraction of the immature heart by AngII. Here we found that AngII significantly increased the peak amplitude of twitch Ca2+ transients by robustly activating L-type CaV 1.2 Ca2+ (CaV 1.2) channels in mouse immature but not mature cardiomyocytes. This response to AngII was mediated by AT1 receptors and ß-arrestin2. A ß-arrestin-biased AT1 receptor agonist was as effective as AngII in activating CaV 1.2 channels. Src-family tyrosine kinases (SFKs) and casein kinase 2α'ß (CK2α'ß) were sequentially activated when AngII activated CaV 1.2 channels. A cyclin-dependent kinase inhibitor, p27Kip1 (p27), inhibited CK2α'ß, and AngII removed this inhibitory effect through phosphorylating tyrosine 88 of p27 via SFKs in cardiomyocytes. In a human embryonic kidney cell line, tsA201 cells, overexpression of CK2α'ß but not c-Src directly activated recombinant CaV 1.2 channels composed of C-terminally truncated α1C , the distal C-terminus of α1C , ß2C and α2 δ1 subunits, by phosphorylating threonine 1704 located at the interface between the proximal and the distal C-terminus of CaV 1.2α1C subunits. Co-immunoprecipitation revealed that CaV 1.2 channels, CK2α'ß and p27 formed a macromolecular complex. Therefore, stimulation of AT1 receptors by AngII activates CaV 1.2 channels through ß-arrestin2 and CK2α'ß, thereby probably exerting a positive inotropic effect in the immature heart. Our results also indicated that ß-arrestin-biased AT1 receptor agonists may be used as valuable therapeutics for paediatric heart failure in the future.


Assuntos
Angiotensina II/farmacologia , Canais de Cálcio Tipo L/metabolismo , Caseína Quinase II/metabolismo , Miócitos Cardíacos/metabolismo , beta-Arrestina 2/metabolismo , Potenciais de Ação , Animais , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Receptor Tipo 1 de Angiotensina/metabolismo
19.
J Mol Cell Cardiol ; 81: 139-49, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25724725

RESUMO

The transplantation of adipose tissue-derived stem cells (ADSCs) improves cardiac contractility after myocardial infarction (MI); however, little is known about the electrophysiological consequences of transplantation. The purpose of this study was to clarify whether the transplantation of ADSCs increases or decreases the incidence of ventricular tachyarrhythmias (VT) in a rat model of MI. MI was induced experimentally by permanent occlusion of the left anterior descending artery of Lewis rats. ADSCs were harvested from GFP-transgenic rats, and were cultured until passage four. ADSCs (10×10(6)) resuspended in 100µL saline or pro-survival cocktail (PSC), which enhances cardiac graft survival, were injected directly into syngeneic rat hearts 1week after MI. The recipients of ADSCs suspended in PSC had a larger graft area compared with those receiving ASDCs suspended in saline at 1week post-transplantation (number of graft cells/section: 148.7±10.6 vs. 22.4±3.4, p<0.05, n=5/group). Thereafter, all ADSC recipients were transplanted with ASDCs in PSC. ADSCs were transplanted into infarcted hearts, and the mechanical and electrophysiological functions were assessed. Echocardiography revealed that ADSC recipients had improved contractile function compared with those receiving PSC vehicle (fractional shortening: 21.1±0.9 vs. 14.1±1.2, p<0.05, n≥12/group). Four weeks post-transplantation, VT was induced via in vivo programmed electrical stimulation. The recipients of ADSCs showed a significantly lower incidence of induced VT compared with the control (31.3% vs. 83.3%, p<0.05, n≥12/group). To understand the electrical activity following transplantation, we performed ex vivo optical mapping using a voltage sensitive dye, and found that ADSC transplantation decreased conduction velocity and its dispersion in the peri-infarct area. These results suggest that ADSC transplantation improved cardiac mechanical and electrophysiological functions in subacute MI.


Assuntos
Adipócitos/fisiologia , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Taquicardia/terapia , Adipócitos/citologia , Animais , Diferenciação Celular , Proliferação de Células , Vasos Coronários/patologia , Modelos Animais de Doenças , Estimulação Elétrica , Sistema de Condução Cardíaco , Masculino , Contração Miocárdica , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Ratos , Ratos Endogâmicos Lew , Células-Tronco/citologia , Taquicardia/patologia , Taquicardia/fisiopatologia , Transplante Isogênico
20.
PLoS One ; 9(8): e106354, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25171374

RESUMO

BACKGROUND: Transgenic mice with transient cardiac expression of constitutively active Galpha q (Gαq-TG) exhibt progressive heart failure and ventricular arrhythmias after the initiating stimulus of transfected constitutively active Gαq becomes undetectable. However, the mechanisms are still unknown. We examined the effects of chronic administration of olmesartan on heart failure and ventricular arrhythmia in Gαq-TG mice. METHODOLOGY/PRINCIPAL FINDINGS: Olmesartan (1 mg/kg/day) or vehicle was chronically administered to Gαq-TG from 6 to 32 weeks of age, and all experiments were performed in mice at the age of 32 weeks. Chronic olmesartan administration prevented the severe reduction of left ventricular fractional shortening, and inhibited ventricular interstitial fibrosis and ventricular myocyte hypertrophy in Gαq-TG. Electrocardiogram demonstrated that premature ventricular contraction (PVC) was frequently (more than 20 beats/min) observed in 9 of 10 vehicle-treated Gαq-TG but in none of 10 olmesartan-treated Gαq-TG. The collected QT interval and monophasic action potential duration in the left ventricle were significantly shorter in olmesartan-treated Gαq-TG than in vehicle-treated Gαq-TG. CTGF, collagen type 1, ANP, BNP, and ß-MHC gene expression was increased and olmesartan significantly decreased the expression of these genes in Gαq-TG mouse ventricles. The expression of canonical transient receptor potential (TRPC) 3 and 6 channel and angiotensin converting enzyme (ACE) proteins but not angiotensin II type 1 (AT1) receptor was increased in Gαq-TG ventricles compared with NTG mouse ventricles. Olmesartan significantly decreased TRPC6 and tended to decrease ACE expressions in Gαq-TG. Moreover, it increased AT1 receptor in Gαq-TG. CONCLUSIONS/SIGNIFICANCE: These findings suggest that angiotensin II type 1 receptor activation plays an important role in the development of heart failure and ventricular arrhythmia in Gαq-TG mouse model of heart failure.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Insuficiência Cardíaca/fisiopatologia , Imidazóis/administração & dosagem , Tetrazóis/administração & dosagem , Complexos Ventriculares Prematuros/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Fator Natriurético Atrial/metabolismo , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/prevenção & controle , Camundongos , Camundongos Transgênicos , Cadeias Pesadas de Miosina/metabolismo , Complexos Ventriculares Prematuros/genética , Complexos Ventriculares Prematuros/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...