Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 20(1): 435, 2022 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-36195937

RESUMO

Exosomes of human cardiosphere-derived cells (CDCs) are very promising for treating cardiovascular disorders. However, the current challenge is inconvenient delivery methods of exosomes for clinical application. The present study aims to explore the potential to enhance the therapeutic effect of exosome (EXO) from human CDCs to myocardial hypertrophy. A heart homing peptide (HHP) was displayed on the surface of exosomes derived from CDCs that were forced to express the HHP fused on the N-terminus of the lysosomal-associated membrane protein 2b (LAMP2b). The cardiomyocyte-targeting capability of exosomes were analyzed and their therapeutic effects were evaluated in a mouse model of myocardial hypertrophy induced by transverse aorta constriction (TAC). The molecular mechanisms of the therapeutic effects were dissected in angiotensin II-induced neonatal rat cardiomyocyte (NRCMs) hypertrophy model using a combination of biochemistry, immunohistochemistry and molecular biology techniques. We found that HHP-exosomes (HHP-EXO) accumulated more in mouse hearts after intravenous delivery and in cultured NRCMs than control exosomes (CON-EXO). Cardiac function of TAC mice was significantly improved with intravenous HHP-EXO administration. Left ventricular hypertrophy was reduced more by HHP-EXO than CON-EXO via inhibition of ß-MHC, BNP, GP130, p-STAT3, p-ERK1/2, and p-AKT. Similar results were obtained in angiotensin II-induced hypertrophy of NRCMs, in which the beneficial effects of HHP-EXO were abolished by miRNA-148a inhibition. Our results indicate that HHP-EXO preferentially target the heart and improve the therapeutic effect of CDCs-exosomes on cardiac hypertrophy. The beneficial therapeutic effect is most likely attributed to miRNA-148a-mediated suppression of GP130, which in turn inhibits STAT3/ERK1/2/AKT signaling pathway, leading to improved cardiac function and remodeling.


Assuntos
Exossomos , MicroRNAs , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Cardiomegalia/terapia , Receptor gp130 de Citocina/metabolismo , Exossomos/metabolismo , Humanos , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 24(9): 1001-1007, 2022.
Artigo em Zh | MEDLINE | ID: mdl-36111718

RESUMO

OBJECTIVES: To study the association between neonatal discharge preparedness and adverse health events. METHODS: The neonates who were born in hospitals from different regions of Gansu Province in China and their parents were enrolled as subjects, and an investigation was performed for the discharge preparedness. According to the level of discharge preparedness, the subjects were divided into low-, middle-, and high-level groups. The neonates were followed up to observe the incidence rate of adverse health events within one month after discharge. The association between neonatal discharge preparedness and adverse health events was analyzed. RESULTS: The neonates with adverse health events had a significantly lower level of discharge preparedness than those without adverse events (P<0.05). The multivariate logistic regression analysis showed that the incidence rate of adverse health events was reduced by 34.8% in the middle-level group and 78.7% in the high-level group compared with the low-level group (P<0.05). The readmission rate of neonates was 8.1% (35/430), and the neonates readmitted had a significantly lower level of discharge preparedness than those not readmitted (P<0.05). The multivariate logistic regression analysis showed that the readmission rate of neonates was reduced by 67.4% in the middle-level group and 84.2% in the high-level group compared with the low-level group (P<0.05). CONCLUSIONS: Discharge preparedness may affect the incidence of adverse health events and the rate of readmission within one month after discharge. Medical staff should adopt effective intervention measures to improve discharge preparedness, so as to reduce the incidence of adverse health events and the rate of readmission.


Assuntos
Alta do Paciente , Readmissão do Paciente , China , Humanos , Incidência , Recém-Nascido
3.
J Cell Mol Med ; 24(20): 12141-12153, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32918384

RESUMO

Doxorubicin cardiotoxicity is frequently reported in patients undergoing chemotherapy. The present study investigates whether cardiomyopathy induced by doxorubicin can be improved by the natural flavone acacetin in a mouse model and uncovers the potential molecular mechanism using cultured rat cardiomyoblasts. It was found that the cardiac dysfunction and myocardial fibrosis induced by doxorubicin were significantly improved by acacetin in mice with impaired Nrf2/HO-1 and Sirt1/pAMPK molecules, which is reversed by acacetin treatment. Doxorubicin decreased cell viability and increased ROS production in rat cardiomyoblasts; these effects are significantly countered by acacetin (0.3-3 µM) in a concentration-dependent manner via activating Sirt1/pAMPK signals and enhancing antioxidation (Nrf2/HO-1 and SOD1/SOD2) and anti-apoptosis. These protective effects were abolished in cells with silencing Sirt1. The results demonstrate for the first time that doxorubicin cardiotoxicity is antagonized by acacetin via Sirt1-mediated activation of AMPK/Nrf2 signal molecules, indicating that acacetin may be a drug candidate used clinically for protecting against doxorubicin cardiomyopathy.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/tratamento farmacológico , Doxorrubicina/efeitos adversos , Flavonas/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Cardiomiopatias/metabolismo , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Flavonas/farmacologia , Inativação Gênica , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Masculino , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Ratos , Espécies Reativas de Oxigênio/metabolismo
4.
J Cell Mol Med ; 22(10): 4688-4699, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30117680

RESUMO

Our recent study showed that bradykinin increases cell cycling progression and migration of human cardiac c-Kit+ progenitor cells by activating pAkt and pERK1/2 signals. This study investigated whether bradykinin-mediated Ca2+ signalling participates in regulating cellular functions in cultured human cardiac c-Kit+ progenitor cells using laser scanning confocal microscopy and biochemical approaches. It was found that bradykinin increased cytosolic free Ca2+ ( Cai2+ ) by triggering a transient Ca2+ release from ER IP3Rs followed by sustained Ca2+ influx through store-operated Ca2+ entry (SOCE) channel. Blockade of B2 receptor with HOE140 or IP3Rs with araguspongin B or silencing IP3R3 with siRNA abolished both Ca2+ release and Ca2+ influx. It is interesting to note that the bradykinin-induced cell cycle progression and migration were not observed in cells with siRNA-silenced IP3R3 or the SOCE component TRPC1, Orai1 or STIM1. Also the bradykinin-induced increase in pAkt and pERK1/2 as well as cyclin D1 was reduced in these cells. These results demonstrate for the first time that bradykinin-mediated increase in free Cai2+ via ER-IP3R3 Ca2+ release followed by Ca2+ influx through SOCE channel plays a crucial role in regulating cell growth and migration via activating pAkt, pERK1/2 and cyclin D1 in human cardiac c-Kit+ progenitor cells.


Assuntos
Bradicinina/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Células-Tronco/efeitos dos fármacos , Cátions Bivalentes , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Transporte de Íons/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/antagonistas & inibidores , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/metabolismo , Quinolizinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Molécula 1 de Interação Estromal/antagonistas & inibidores , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo
5.
J Cell Mol Med ; 21(9): 1826-1834, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28294531

RESUMO

The present study was designed to investigate whether large conductance Ca2+ -activated K+ (BK) channels were regulated by epidermal growth factor (EGF) receptor (EGFR) tyrosine kinase. BK current and channel tyrosine phosphorylation level were measured in BK-HEK 293 cells expressing both functional α-subunits and the auxiliary ß1-subunits using electrophysiology, immunoprecipitation and Western blotting approaches, respectively, and the function of rat cerebral basilar arteries was determined with a wire myography system. We found that BK current in BK-HEK 293 cells was increased by the broad spectrum protein tyrosine kinase (PTK) inhibitor genistein and the selective EGFR tyrosine kinase inhibitor AG556, one of the known tyrphostin. The effect of genistein or AG556 was antagonized by the protein tyrosine phosphatase (PTP) inhibitor orthovanadate. On the other hand, orthovanadate or EGF decreased BK current, and the effect was counteracted by AG556. The tyrosine phosphorylation level of BK channels (α- and ß1-subunits) was increased by EGF and orthovanadate, while decreased by genistein and AG556, and the reduced tyrosine phosphorylation of BK channels by genistein or AG556 was reversed by orthovanadate. Interestingly, AG556 induced a remarkable enhancement of BK current in rat cerebral artery smooth muscle cells and relaxation of pre-contracted rat cerebral basilar arteries with denuded endothelium, and these effects were antagonized by the BK channel blocker paxilline or orthovanadate. These results demonstrate that tyrosine phosphorylation of BK channels by EGFR kinase decreases the channel activity, and inhibition of EGFR kinase by AG556 enhances the channel activity and dilates rat cerebral basilar arteries.


Assuntos
Receptores ErbB/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Tirfostinas/farmacologia , Animais , Artéria Basilar/citologia , Separação Celular , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Genisteína/farmacologia , Células HEK293 , Humanos , Indóis/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Subunidades Proteicas/metabolismo , Ratos Sprague-Dawley , Vanadatos/farmacologia , Vasodilatação/efeitos dos fármacos
6.
J Cell Mol Med ; 20(6): 1118-27, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26865051

RESUMO

The cellular physiology and biology of human cardiac c-kit(+) progenitor cells has not been extensively characterized and remains an area of active research. This study investigates the functional expression of transient receptor potential vanilloid (TRPV) and possible roles for this ion channel in regulating proliferation and migration of human cardiac c-kit(+) progenitor cells. We found that genes coding for TRPV2 and TRPV4 channels and their proteins are significantly expressed in human c-kit(+) cardiac stem cells. Probenecid, an activator of TRPV2, induced an increase in intracellular Ca(2+) (Ca(2+) i ), an effect that may be attenuated or abolished by the TRPV2 blocker ruthenium red. The TRPV4 channel activator 4α-phorbol 12-13-dicaprinate induced Ca(2+) i oscillations, which can be inhibited by the TRPV4 blocker RN-1734. The alteration of Ca(2+) i by probenecid or 4α-phorbol 12-13-dicprinate was dramatically inhibited in cells infected with TRPV2 short hairpin RNA (shRNA) or TRPV4 shRNA. Silencing TRPV2, but not TRPV4, significantly reduced cell proliferation by arresting cells at the G0/G1 boundary of the cell cycle. Cell migration was reduced by silencing TRPV2 or TRPV4. Western blot revealed that silencing TRPV2 decreased expression of cyclin D1, cyclin E, pERK1/2 and pAkt, whereas silencing TRPV4 only reduced pAkt expression. Our results demonstrate for the first time that functional TRPV2 and TRPV4 channels are abundantly expressed in human cardiac c-kit(+) progenitor cells. TRPV2 channels, but not TRPV4 channels, participate in regulating cell cycle progression; moreover, both TRPV2 and TRPV4 are involved in migration of human cardiac c-kit(+) progenitor cells.


Assuntos
Miocárdio/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/metabolismo , Canais de Cátion TRPV/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , Movimento Celular , Proliferação de Células , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Espaço Intracelular/metabolismo , RNA Interferente Pequeno/metabolismo , Células-Tronco/citologia , Canais de Cátion TRPV/genética
7.
Pharmacol Res ; 107: 186-194, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26995303

RESUMO

The present study was designed to investigate the effect of equol on cerebral blood flow and the underlying molecular mechanisms. The regional cerebral blood flow in parietal lobe of rats was measured by using a laser Doppler flowmetry. Isolated cerebral basilar artery and mesenteric artery rings from rats were used for vascular reactivity measurement with a multi wire myography system. Outward K(+) current in smooth muscle cells of cerebral basilar artery, large-conductance Ca(2+)-activated K(+) (BK) channel current in BK-HEK 293 cells stably expressing both human α (hSlo)- and ß1-subunits, and hSlo channel current in hSlo-HEK 293 cells expressing only the α-subunit of BK channels were recorded with whole cell patch-clamp technique. The results showed that equol significantly increased regional cerebral blood flow in rats, and produced a concentration-dependent but endothelium-independent relaxation in rat cerebral basilar arteries. Both paxilline and iberiotoxin, two selective BK channel blockers, significantly inhibited equol-induced vasodilation in cerebral arteries. Outward K(+) currents in smooth muscle cells of cerebral basilar artery were increased by equol and fully reversed by washout or blockade of BK channels with iberiotoxin. Equol remarkably enhanced human BK current in BK-HEK 293 cells, but not hSlo current in hSlo-HEK 293 cells, and the increase was completely abolished by co-application of paxilline. Our findings provide the first information that equol selectively stimulates BK channel current by acting on its ß1 subunit, which may in turn contribute to the equol-mediated vasodilation and cerebral blood flow increase.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Equol/farmacologia , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/fisiologia , Células HEK293 , Humanos , Técnicas In Vitro , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Ratos , Ratos Sprague-Dawley , Vasodilatação/efeitos dos fármacos
8.
Pharmacol Res ; 104: 61-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26689773

RESUMO

SKF-96365 is a TRPC channel antagonist commonly used to characterize the potential functions of TRPC channels in different systems, which was recently reported to induce QTc prolongation on ECG by inhibiting TRPC channels. The present study investigates whether the blockade of cardiac repolarization currents would be involved in the increase of QTc interval. Cardiac repolarization currents were recorded in HEK 293 cells stably expressing human ether-à-go-go-related gene potassium (hERG or hKv11.1) channels, hKCNQ1/hKCNE1 channels (IKs) or hKir2.1 channels and cardiac action potentials were recorded in guinea pig ventricular myocytes using a whole-cell patch technique. The potential effect of SKF-96365 on QT interval was evaluated in ex vivo guinea pig hearts. It was found that SKF-96365 inhibited hERG current in a concentration-dependent manner (IC50, 3.4µM). The hERG mutants S631A in the pore helix and F656V of the S6 region reduced the inhibitory sensitivity with IC50s of 27.4µM and 11.0µM, suggesting a channel pore blocker. In addition, this compound inhibited IKs and hKir2.1currents with IC50s of 10.8 and 8.7µM. SKF-96365 (10µM) significantly prolonged ventricular APD90 in guinea pig ventricular myocytes and QTc interval in ex vivo guinea pig hearts. These results indicate that the TRPC channel antagonist SKF-96365 exerts blocking effects on hERG, IKs, and hKir2.1 channels. Prolongation of ventricular APD and QT interval is related to the inhibition of multiple repolarization potassium currents, especially hERG channels.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Imidazóis/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Animais , Eletrocardiografia/efeitos dos fármacos , Cobaias , Células HEK293 , Coração/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia
9.
Pflugers Arch ; 467(6): 1227-36, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25017106

RESUMO

SKF-96365 (1-(beta-[3-(4-methoxy-phenyl) propoxy]-4-methoxyphenethyl)-1H-imidazole hydrochloride) is a general TRPC channel antagonist commonly used to characterize the potential functions of TRPC channels in cardiovascular system. Recent reports showed that SKF-96365 induced a reduction in cardiac conduction. The present study investigates whether the reduced cardiac conduction caused by SKF-96365 is related to the blockade of voltage-gated sodium current (I Na) in rat ventricular myocytes using the whole-cell patch voltage-clamp technique. It was found that SKF-96365 inhibited I Na in rat ventricular myocytes in a concentration-dependent manner. The compound (1 µM) negatively shifted the potential of I Na availability by 9.5 mV, increased the closed-state inactivation of I Na, and slowed the recovery of I Na from inactivation. The inhibition of cardiac I Na by SKF-96365 was use-dependent and frequency-dependent, and the IC50 was decreased from 1.36 µM at 0.5 Hz to 1.03, 0.81, 0.61, 0.56 µM at 1, 2, 5, 10 Hz, respectively. However, the selective TRPC3 antagonist Pyr3 decreased cardiac I Na by 8.5% at 10 µM with a weak use and frequency dependence. These results demonstrate that the TRPC channel antagonist SKF-96365 strongly blocks cardiac I Na in use-dependent and frequency-dependent manners. Caution should be taken for interpreting the alteration of cardiac electrical activity when SKF-96365 is used in native cells as a TRPC antagonist.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Ventrículos do Coração/citologia , Imidazóis/farmacologia , Miócitos Cardíacos/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Células Cultivadas , Células HEK293 , Humanos , Concentração Inibidora 50 , Miócitos Cardíacos/efeitos dos fármacos , Pirazóis/farmacologia , Ratos
10.
Am J Physiol Heart Circ Physiol ; 309(10): H1772-81, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26453325

RESUMO

Cardiac c-kit(+) progenitor cells are important for maintaining cardiac homeostasis and can potentially contribute to myocardial repair. However, cellular physiology of human cardiac c-kit(+) progenitor cells is not well understood. The present study investigates the functional store-operated Ca(2+) entry (SOCE) channels and the potential role in regulating cell cycling and migration using confocal microscopy, RT-PCR, Western blot, coimmunoprecipitation, cell proliferation, and migration assays. We found that SOCE channels mediated Ca(2+) influx, and TRPC1, STIM1, and Orai1 were involved in the formation of SOCE channels in human cardiac c-kit(+) progenitor cells. Silencing TRPC1, STIM1, or Orai1 with the corresponding siRNA significantly reduced the Ca(2+) signaling through SOCE channels, decreased cell proliferation and migration, and reduced expression of cyclin D1, cyclin E, and/or p-Akt. Our results demonstrate the novel information that Ca(2+) signaling through SOCE channels regulates cell cycling and migration via activating cyclin D1, cyclin E, and/or p-Akt in human cardiac c-kit(+) cells.


Assuntos
Canais de Cálcio/genética , Sinalização do Cálcio/genética , Ciclo Celular/genética , Movimento Celular/genética , Miocárdio/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/metabolismo , Ensaios de Migração Celular , Proliferação de Células/genética , Células Cultivadas , Ciclina D1/metabolismo , Ciclina E/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Imunoprecipitação , Proteínas de Membrana/genética , Microscopia Confocal , Proteínas de Neoplasias/genética , Proteína ORAI1 , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/genética
11.
Anesthesiology ; 122(3): 571-84, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25321870

RESUMO

BACKGROUND: Propofol is widely used clinically for the induction and maintenance of anesthesia. Clinical case reports have shown that propofol has an antiatrial tachycardia/fibrillation effect; however, the related ionic mechanisms are not fully understood. The current study investigates the effects of propofol on human cardiac potassium channels. METHODS: The whole cell patch voltage clamp technique was used to record transient outward potassium current (Ito) and ultrarapidly activating delayed rectifier potassium current (IKur) in human atrial myocytes and hKv1.5, human ether-à-go-go-related gene (hERG), and hKCNQ1/hKCNE1 channels stably expressed in HEK 293 cells. Current clamp mode was used to record action potentials in human atrial myocytes. RESULTS: In human atrial myocytes, propofol inhibited Ito in a concentration-dependent manner (IC50 = 33.5 ± 2.0 µM for peak current, n = 6) by blocking open channels without affecting the voltage-dependent kinetics or the recovery time constant; propofol decreased IKur (IC50 = 35.3 ± 1.9 µM, n = 6) in human atrial myocytes and inhibited hKv1.5 current expressed in HEK 293 cells by preferentially binding to the open channels. Action potential duration at 90% repolarization was slightly prolonged by 30 µM propofol in human atrial myocytes. In addition, propofol also suppressed hERG and hKCNQ1/hKCNE1 channels expressed in HEK 293 cells. CONCLUSION: Propofol inhibits multiple human cardiac potassium channels, including human atrial Ito and IKur, as well as hKv1.5, hERG, and hKCNQ1/hKCNE1 channels stably expressed in HEK 293 cells, and slightly prolongs human atrial action potential duration, which may contribute to the antiatrial tachycardia/fibrillation effects observed in patients who receive propofol.


Assuntos
Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Bloqueadores dos Canais de Potássio/administração & dosagem , Canais de Potássio/fisiologia , Propofol/administração & dosagem , Idoso , Anestésicos Intravenosos , Células Cultivadas , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade
12.
J Cell Physiol ; 229(1): 60-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23765921

RESUMO

Transient receptor potential melastatin-7 (TRPM7) channels are involved in many cellular physiological and pathological processes. The present study was designed to investigate the expression of TRPM7 channels and the potential role in regulating cell proliferation and adipogenesis in 3T3-L1 preadipocytes with approaches of whole-cell patch voltage-clamp, molecular biology, cell proliferation, adipogenesis, etc. We found that a TRPM7-like current was recorded with Mg(2+) -free pipette solution in 3T3-L1 preadipocytes, and the current was inhibited by intercellular free Mg(2+) . The TRPM7-like current was potentiated by acidic pH and inhibited by 2-aminoethoxydiphenyl borate (2-APB). RT-PCR, Western blot and immunocytochemistry revealed that gene and protein of TRPM7 channels were abundant in 3T3-L1 preadipocytes. Blockade of TRPM7 channels with 2-APB inhibited cell proliferation in 3T3-L1 cells. In addition, knockdown of TRPM7 with specific siRNA inhibited both proliferation and adipogenesis. The present study demonstrates for the first time that TRPM7 channels regulate cell cycle and adipogenesis of 3T3-L1 preadipocytes.


Assuntos
Adipogenia/genética , Canais de Cátion TRPM/genética , Células 3T3-L1 , Animais , Compostos de Boro/farmacologia , Proliferação de Células/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Camundongos , Técnicas de Patch-Clamp , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/biossíntese
13.
J Cell Physiol ; 229(2): 202-12, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23881642

RESUMO

Human bone marrow-derived mesenchymal stem cells (MSCs) serve as a reservoir for the continuous renewal of various mesenchymal tissues; however, cellular physiology of ion channels is not fully understood. The present study investigated potential roles of large-conductance Ca(2+) -activated potassium (BKCa ) channels and ether-à-go-go potassium (hEag1 or Kv10.1) channels in regulating cell proliferation and differentiation in human MSCs. We found that inhibition of BKCa with paxilline or hEag1 with astemizole, or knockdown of BKCa with shRNAs targeting KCa1.1 or hEag1 channels with shRNAs targeting KCNH1 arrested the cells at G0/G1 phase. In addition, silencing BKCa or hEag1 channels significantly reduced adipogenic differentiation with decrease of lipid accumulation and expression of the adipocyte marker PPARγ, and decreased osteogenic differentiation with reduction of mineral precipitation and osteocalcin. These effects were accompanied with a reduced cyclin D1, cyclin E, p-ERK1/2, and p-Akt. Our results demonstrate that BKCa and hEag1 channels not only regulate cell proliferation, but also participate in the adipogenic and osteogenic differentiations in human MSCs, which indicates that BKCa and hEag1 channels may be essential in maintaining bone marrow physiological function and bone regeneration.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Proliferação de Células , Canais de Potássio Éter-A-Go-Go/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Células-Tronco Mesenquimais/citologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Indóis/farmacologia , Ativação do Canal Iônico , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/antagonistas & inibidores , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Células-Tronco Mesenquimais/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno
14.
Pflugers Arch ; 466(5): 947-59, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24057349

RESUMO

Preadipocytes are widely used as an in vitro model to investigate proliferation, adipogenic differentiation, and lipodystrophy; however, cellular physiology and biology are not fully understood in human preadipocytes. The present study was to investigate the expression of transient receptor potential (TRP) channels in human preadipocytes and their potential roles in regulating proliferation and adipogenic differentiation using approaches of confocal microscopy, whole-cell patch voltage-clamp, reverse transcription polymerase chain reaction, Western blot, etc. We found that TRPV2, TRPV4, and TRPM7 channels were abundantly expressed in human preadipocytes. The intracellular Ca(2+) transient activated by the TRPV2 activator probenecid was reversed or prevented by ruthenium red, a TRPV2 blocker. The TRPV4 channel activator, 4α-phorbol 12-13-dicaprinate, enhanced intracellular Ca(2+) oscillations, and the effect was inhibited by the TRPV4 blocker RN-1734. TRPM7 current was recorded with dialysis of Mg(2+)-free pipette solution, which was inhibited by the TRP channel blocker 2-aminoethoxydiphenyl borate and enhanced by acidic extracellular pH. Silencing TRPV2 or TRPM7, but not TRPV4, significantly reduced cell proliferation via inhibiting cyclin D1, cyclin E, and p-ERK1/2. Interestingly, individually silencing these three channels decreased adipogenic differentiation of human preadipocytes by reducing p-Akt kinase. Our results demonstrate for the first time that functional TRPV2, TRPV4, and TRPM7 channels are abundantly expressed in human preadipocytes. TRPV2 and TRPM7, but not TRPV4, regulate cell proliferation via activating cyclin D1, cyclin E, and p-ERK1/2, while they are all involved in adipogenesis in human preadipocytes via phosphorylating Akt kinase.


Assuntos
Potenciais de Ação , Adipócitos/metabolismo , Células-Tronco Adultas/metabolismo , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/metabolismo , Adipócitos/citologia , Adipócitos/fisiologia , Adipogenia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Sinalização do Cálcio , Proliferação de Células , Ciclina D1/metabolismo , Ciclina E/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Ésteres de Forbol/farmacologia , Probenecid/farmacologia , Proteínas Serina-Treonina Quinases , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rutênio Vermelho/farmacologia , Sulfonamidas/farmacologia , Canais de Cátion TRPM/genética , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética
15.
Basic Res Cardiol ; 109(3): 407, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24691761

RESUMO

Cardiac progenitor cells play an important role in cardiac repair and regeneration; however, their cellular biology and electrophysiology are not understood. The present study characterizes the functional ion channels in human cardiac c-kit(+) progenitor cells using whole-cell patch voltage-clamp, RT-PCR, and Western blots. We found that several ionic currents were present in human cardiac c-kit(+) progenitor cells, including a large-conductance Ca(2+)-activated K(+) current (BKCa) in 86 % of cells, an inwardly rectifying K(+) current (I Kir) in 84 % of cells, a transient outward K(+) current (I to) in 47 % of cells, a voltage-gated tetrodotoxin-sensitive Na(+) current (I Na,TTX) in 61 % of cells. Molecular identities of these ionic currents were determined with RT-PCR and Western-blot analysis. KCa.1.1 (for BKCa), Kir2.1 (for I Kir), Kv4.2 and Kv4.3 (for I to), Nav1.3 and Nav1.6 (for I Na.TTX) were abundantly expressed in human cardiac c-kit(+) progenitor cells, which do not resemble cardiomyocytes at all. These results demonstrate for the first time that four types of ionic currents including BKCa, I to, I Kir, and I Na.TTX, are heterogeneously present in human cardiac c-kit(+) cells, which may be involved in regulating cellular physiology.


Assuntos
Canais Iônicos/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/metabolismo , Idoso , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Feminino , Humanos , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/genética , Masculino , Potenciais da Membrana , Moduladores de Transporte de Membrana/farmacologia , Pessoa de Meia-Idade , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/efeitos dos fármacos , Fatores de Tempo
16.
Biochem J ; 452(1): 121-9, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23496660

RESUMO

SKCa (small-conductance Ca(2+)-activated K(+)) channels are widely distributed in different tissues, including the brain, pancreatic islets and myocardium and play an important role in controlling electrical activity and cellular functions. However, intracellular signal modulation of SKCa channels is not fully understood. The present study was designed to investigate the potential regulation of hSKCa1 (human SKCa1) channels by PTKs (protein tyrosine kinases) in HEK (human embryonic kidney)-293 cells expressing the hSKCa1 (KCNN1) gene using approaches of whole-cell patch voltage-clamp, immunoprecipitation, Western blotting and mutagenesis. We found that the hSKCa1 current was inhibited by the broad-spectrum PTK inhibitor genistein, the selective EGFR (epidermal growth factor receptor) kinase inhibitors T25 (tyrphostin 25) and AG556 (tyrphostin AG 556), but not by the Src-family kinases inhibitor PP2. The inhibitory effect of these PTK inhibitors was significantly antagonized by the PTP (protein tyrosine phosphatase) inhibitor orthovanadate. The tyrosine phosphorylation level of hSKCa1 channels was reduced by genistein, T25 or AG556. The reduced tyrosine phosphorylation was countered by orthovanadate. Interestingly, the Y109F mutant hSKCa1 channel lost the inhibitory response to T25 or AG556, and showed a dramatic reduction in tyrosine phosphorylation levels and a reduced current density. These results demonstrate the novel information that hSKCa1 channels are inhibited by genistein, T25 and AG556 via EGFR tyrosine kinase inhibition, which is related to the phosphorylation of Tyr(109) in the N-terminus. This effect may affect electrical activity and cellular functions in brain, pancreatic islets and myocardium.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Genisteína/farmacologia , Células HEK293 , Humanos , Inibidores de Proteínas Quinases/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/biossíntese , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Tirfostinas/farmacologia
17.
Sheng Li Xue Bao ; 66(1): 85-95, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24553873

RESUMO

Cellular excitability is an important physiological factor in maintaining normal cardiac activity. The present study was designed to investigate the ionic mechanism underlying different excitability in atrial and ventricular myocytes of guinea pig heart using a whole-cell patch configuration. We found that excitability is lower in ventricular myocytes than that in atrial myocytes. Although the density of voltage-gated fast Na(+) current (INa) was lower in ventricular myocytes, it would not correlate to the lower excitability since its availability was greater than that in atrial myocytes around threshold potential. Classical inward rectifier K(+) current (IK1) was greater in ventricular myocytes than that in atrial myocytes, which might contribute in part to the lower excitability. In addition, the transient outward K(+) current with inward rectification (Itoir) elicited by depolarization was greater in ventricular myocytes than that in atrial myocytes and might contribute to the lower excitability. In ventricular myocytes, Ba(2+) at 5 µmol/L significantly inhibited Itoir, enhanced excitability, and shifted the threshold potential of INa activation to more negative, and the effect was independent of affecting INa. Our results demonstrate the novel information that in addition to classical IK1, Itoir plays a major role in determining the distinctive excitability in guinea pig atrial and ventricular myocytes and maintaining cardiac excitability. More effort is required to investigate whether increase of Itoir would be protective via reducing excitability.


Assuntos
Função Atrial , Miócitos Cardíacos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Função Ventricular , Canais de Sódio Disparados por Voltagem/fisiologia , Animais , Cobaias , Átrios do Coração/citologia , Ventrículos do Coração/citologia
18.
Chin Med ; 19(1): 68, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741130

RESUMO

BACKGROUND: Myocarditis refers to an autoimmune inflammatory response of the myocardium with characterization of self-reactive CD4+ T cell activation, which lacks effective treatment and has a poor prognosis. Acacetin is a natural flavonoid product that has been reported to have anti-inflammatory effects. However, acacetin has not been investigated in myocarditis. METHODS: Oral acacetin treatment was administered in an experimental autoimmune myocarditis model established with myosin heavy chain-alpha peptide. Echocardiography, pathological staining, and RT-qPCR were used to detect cardiac function, myocardial injury, and inflammation levels. Flow cytometry was utilized to detect the effect of acacetin on CD4+ T cell function. RNA-seq, molecular docking, and microscale thermophoresis (MST) were employed to investigate potential mechanisms. Seahorse analysis, mitoSOX, JC-1, and mitotracker were utilized to detect the effect of acacetin on mitochondrial function. RESULTS: Acacetin attenuated cardiac injury and fibrosis as well as heart dysfunction, and reduced cardiac inflammatory cytokines and ratio of effector CD4+ T and Th17 cells. Acacetin inhibited CD4+ T cell activation, proliferation, and Th17 cell differentiation. Mechanistically, the effects of acacetin were related to reducing mitochondrial complex II activity thereby inhibiting mitochondrial respiration and mitochondrial reactive oxygen species in CD4+ T cells. CONCLUSION: Acacetin may be a valuable therapeutic drug in treating CD4+ T cell-mediated myocarditis.

19.
J Biol Chem ; 287(42): 35599-35611, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22908234

RESUMO

Cyclic adenosine diphosphoribose (cADPR) is an endogenous Ca(2+) mobilizing messenger that is formed by ADP-ribosyl cyclases from nicotinamide adenine dinucleotide (NAD). The main ADP-ribosyl cyclase in mammals is CD38, a multi-functional enzyme and a type II membrane protein. Here we explored the role of CD38-cADPR-Ca(2+) in the cardiomyogenesis of mouse embryonic stem (ES) cells. We found that the mouse ES cells are responsive to cADPR and possess the key components of the cADPR signaling pathway. In vitro cardiomyocyte (CM) differentiation of mouse ES cells was initiated by embryoid body (EB) formation. Interestingly, beating cells appeared earlier and were more abundant in CD38 knockdown EBs than in control EBs. Real-time RT-PCR and Western blot analyses further showed that the expression of several cardiac markers, including GATA4, MEF2C, NKX2.5, and α-MLC, were increased markedly in CD38 knockdown EBs than those in control EBs. Similarly, FACS analysis showed that more cardiac Troponin T-positive CMs existed in CD38 knockdown or 8-Br-cADPR, a cADPR antagonist, treated EBs compared with that in control EBs. On the other hand, overexpression of CD38 in mouse ES cells significantly inhibited CM differentiation. Moreover, CD38 knockdown ES cell-derived CMs possess the functional properties characteristic of normal ES cell-derived CMs. Last, we showed that the CD38-cADPR pathway negatively modulated the FGF4-Erks1/2 cascade during CM differentiation of ES cells, and transiently inhibition of Erk1/2 blocked the enhanced effects of CD38 knockdown on the differentiation of CM from ES cells. Taken together, our data indicate that the CD38-cADPR-Ca(2+) signaling pathway antagonizes the CM differentiation of mouse ES cells.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Sinalização do Cálcio/fisiologia , Diferenciação Celular/fisiologia , ADP-Ribose Cíclica/metabolismo , Células-Tronco Embrionárias/enzimologia , Glicoproteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/enzimologia , ADP-Ribosil Ciclase 1/genética , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Linhagem Celular , ADP-Ribose Cíclica/genética , Corpos Embrioides/citologia , Corpos Embrioides/enzimologia , Células-Tronco Embrionárias/citologia , Fator 4 de Crescimento de Fibroblastos/biossíntese , Fator 4 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Glicoproteínas de Membrana/genética , Camundongos , Proteínas Musculares/genética , Miócitos Cardíacos/citologia
20.
Biochim Biophys Acta ; 1823(2): 282-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22061963

RESUMO

Human ether á-go-go gene potassium channels (hEAG1 or Kv10.1) are expressed in brain and various human cancers and play a role in neuronal excitement and tumor progression. However, the functional regulation of hEAG channels by signal transduction is not fully understood. The present study was therefore designed to investigate whether hEAG1 channels are regulated by protein tyrosine kinases (PTKs) in HEK 293 cells stably expressing hEAG1 gene using whole-cell patch voltage-clamp, immunoprecipitation, Western blot, and mutagenesis approaches. We found that the selective epidermal growth factor receptor (EGFR) kinase inhibitor AG556 (10 µM), but not the platelet growth factor receptor (PDGFR) kinase inhibitor AG1295 (10 µM) or the Src-family inhibitor PP2 (10 µM), can inhibit hEAG1 current, and the inhibitory effect can be reversed by the protein tyrosine phosphatase (PTP) inhibitor orthovanadate. Immunoprecipitation and Western blot analysis revealed that tyrosine phosphorylation level of hEAG1 channels was reduced by AG556, and the reduction was significantly countered by orthovanadate. The hEAG1 mutants Y90A, Y344A and Y485A, but not Y376A and Y479A, exhibited reduced response to AG556. Interestingly, the inhibition effect of AG556 was lost in triple mutant hEAG1 channels at Y90, Y344, and Y485 with alanine. These results demonstrate for the first time that hEAG1 channel activity is regulated by EGFR kinase at the tyrosine residues Tyr90, Try344, and Try485. This effect is likely involved in regulating neuronal activity and/or tumor growth.


Assuntos
Receptores ErbB/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Técnicas de Patch-Clamp , Interferência de RNA , Transdução de Sinais/fisiologia , Tirosina/metabolismo , Tirfostinas/metabolismo , Vanadatos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA