Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Ann Surg Oncol ; 2022 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-35192156

RESUMO

BACKGROUND: A potentially curative hepatic resection is the optimal treatment for hepatocellular carcinoma (HCC), but most HCCs, even at an early stage, eventually recur after resection. This study investigates clinical features of initial recurrence and long-term prognosis of patients with recurrence after curative resection for early-stage HCC. PATIENTS AND METHODS: From a multicenter database, patients who underwent curative hepatic resection for early-stage HCC [Barcelona Clinic Liver Cancer (BCLC) stage 0/A] were extracted. Time to initial recurrence, patterns of initial recurrence, and treatment modalities for recurrent tumors were investigated. Univariate and multivariate analysis were used to identify independent risks associated with postoperative recurrence, as well as post-recurrence survival (PRS) for patients with recurrence. RESULTS: Among 1424 patients, 679 (47.7%) developed recurrence at a median follow-up of 54.8 months, including 408 (60.1%) early recurrence (≤ 2 years after surgery) and 271 (39.9%) late recurrence (> 2 years). Independent risks of postoperative recurrence included cirrhosis, preoperative alpha-fetoprotein level > 400 ug/L, tumor size > 5 cm, multiple tumors, satellites, microvascular invasion, and intraoperative blood transfusion. Multivariate analysis revealed that receiving irregular recurrence surveillance, initial tumor beyond Milan criteria, early recurrence, BCLC stage B/C of the recurrent tumor, and noncurative treatments were independently associated with poorer PRS. CONCLUSIONS: Nearly half of patients with early-stage HCC experienced recurrence after resection. Understanding recurrence risks may help identify patients at high risk of recurrence who may benefit from future adjuvant therapies. Meaningful survival even after recurrence can still be achieved by postoperative regular surveillance and curative treatment.

2.
Sleep Breath ; 25(1): 9-16, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32399702

RESUMO

PURPOSE: The pathogenesis of cardiovascular disease (CVD) in patients with obstructive sleep apnea (OSA) is unclear. Several studies have suggested that CVD may be caused by oxidative stress from chronic intermittent hypoxia and associated vascular endothelial dysfunction. Oxidative stress in patients with OSA can induce endothelial cell apoptosis, aggravate vascular endothelial damage, and promote the expression of redox-sensitive genes and adhesion molecules. No meta-analysis has explored whether or not OSA is related to nitric oxide (NO). METHOD: To assess the association between serum/plasma NO levels and OSA, we performed a meta-analysis of the literature on the subject to grade the strength of evidence. RESULTS: OSA was significantly related to decreased serum or plasma NO levels (WMD = - 11.66, 95% CI - 17.21 to - 6.11; P < 0.01). Among the studies analyzed, there was high degree of heterogeneity (I2 = 79%, P < 0.01). Sensitivity analysis showed that after omitting any single study or converting a random effects model (REM) to a fixed effects model (FEM), the main results still held. CONCLUSIONS: This meta-analysis suggests a strong correlation between OSA and serum or plasma NO levels which may explain the link between intermittent hypoxia of OSA and risk of CVD. The strength of this finding may spur further basic and clinical research into vascular endothelial dysfunction in patients with OSA.


Assuntos
Óxido Nítrico/sangue , Apneia Obstrutiva do Sono/sangue , Humanos
3.
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
5.
ORL J Otorhinolaryngol Relat Spec ; 79(3): 153-163, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28407635

RESUMO

BACKGROUND/AIMS: According to recent studies, central auditory impairments are closely related to neurodegenerative diseases. However, the mechanism of central presbycusis remains unclear. Ubiquitin C-terminal hydrolase L1 (UCHL1) is important in maintaining proteasomal activity; however, the detailed mechanism has not yet been fully elucidated. This study aims to investigate the molecular alterations involved in UCHL1 regulation during auditory cortex aging. METHODS: D-Galactose (D-gal) induces oxidative stress and senescence in the auditory cortex, as reported in our previous studies. Primary auditory cortex cells were treated with D-gal for 72 h or 5 days. The proteins related to the ubiquitin proteasome system (UPS) and proteasomal activities were evaluated. UCHL1 was overexpressed, and the effects of UCHL1 on the UPS and proteasomal activity were analyzed. RESULTS: Proteasomal activity was elevated at 72 h and decreased at 5 days in D-gal-treated primary auditory cortex cells. We also found that overexpression of UCHL1 increased the UPS-related proteins UBE1, PSMA7, ubiquitinated proteins, and monoubiquitin, and proteasomal activity. CONCLUSION: The results suggest that UCHL1 may modify the aging process in the auditory cortex by regulating UPS- related proteins.


Assuntos
Envelhecimento/metabolismo , Córtex Auditivo/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina Tiolesterase/metabolismo , Análise de Variância , Animais , Biomarcadores/análise , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade
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.
Biol Blood Marrow Transplant ; 22(2): 212-219, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26555814

RESUMO

Numerous previous studies have suggested that cytotoxic T lymphocyte antigen-4 (CTLA-4) plays an important role in acute graft-versus-host disease (GVHD). How CTLA-4 acts in regulating acute GVHD remains unknown, however. In the present study, we found that, compared with healthy controls, CTLA-4 plasma and relative mRNA levels in patients with acute GVHD were initially decreased and then markedly elevated after 28 days of treatment. CTLA-4 levels were higher in patients with grade I-II acute GVHD compared with those with grade III-IV acute GVHD both before and after treatment. Up-regulation of CTLA-4 significantly increased the luciferase activity and degree of phosphorylation of signal transducer and activator of transcription 3 (STAT3). Meanwhile, T cell activation was significantly inhibited, and levels of IFN-γ, IL-17, and IL-22 decreased. These findings suggest that CTLA-4 might be involved in the pathogenesis of acute GVHD, and may down-regulate T helper 1 cells by increasing STAT3 expression in acute GVHD.


Assuntos
Antígeno CTLA-4/metabolismo , Doença Enxerto-Hospedeiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Adolescente , Adulto , Feminino , Humanos , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Transdução de Sinais , Adulto Jovem
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.
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
10.
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
11.
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
12.
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
13.
J Thromb Thrombolysis ; 38(4): 439-46, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24844803

RESUMO

Monocyte subsets and monocyte-platelet aggregates (MPAs) play important role in atherosclerosis and thrombosis. We aimed to determine their changes in patients with unstable angina (UA). In this cross-sectional case-control study, Global Registry of Acute Coronary Events (GRACE) score was determined in 95 UA patients without elevated troponin level. Thirty age-and-sex matched stable coronary heart disease (CHD) subjects served as control group. The classical (CD14++CD16-, Mon1), the intermediate (CD14++CD16+, Mon2) and the non-classical (CD14+CD16++, Mon3) monocytes, as well as subset-specific MPAs, were measured by flow cytometry. Compared with stable CHD patients, UA patients had increased Mon2 and Mon3 counts (all P < 0.001). For UA subjects, compared with GRACE score-determined low risk patients (GRACE score ≤108, n = 70), intermediate-to-high risk patients (GRACE score >108, n = 25) had higher counts of Mon2 and total MPAs, as well as Mon1- and Mon2-associated MPAs (all P < 0.001). Adjusted binary logistic regression analysis revealed that increased counts of Mon2 subset (for per 5 cells/µL increase, OR 1.186, 95% CI 1.044-1.347, P = 0.009), Mon2 MPAs (for per 5 cells/µL increase, OR 1.228, 95% CI 1.062-1.421, P = 0.006) and total MPAs (for per 5 cells/µL increase, OR 1.072, 95 % CI 1.010-1.137, P = 0.022) independently associated with GRACE score-determined intermediate-to-high risk UA patients. In UA patients with intermediate-to-high risk (determined by GRACE score), counts of Mon2 subset, Mon2-associated MPAs and total MPAs are increased, which are independent of traditional risk factors.


Assuntos
Angina Instável/sangue , Angina Instável/diagnóstico , Plaquetas/metabolismo , Adesão Celular/fisiologia , Monócitos/metabolismo , Agregação Plaquetária/fisiologia , Idoso , Estudos de Casos e Controles , Estudos Transversais , Feminino , Citometria de Fluxo/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Função Plaquetária/métodos
14.
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
15.
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
17.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(2): 561-567, 2024 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-38660867

RESUMO

OBJECTIVE: To explore the clinical characteristics and risk factors of cytomegalovirus(CMV) and Epstein-Barr virus(EBV) co-reactivation after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and its influence on prognosis. METHODS: The clinical data of 222 patients who received allo-HSCT from January 2015 to December 2020 were collected, and the patients were divided into groups according to the occurrence of CMV and EBV infection. Kaplan-Meier method was used for survival analysis, and Cox proportional hazard regression model was used to analyze the risk factors of co-reactivation of CMV and EBV. RESULTS: After allo-HSCT, there were 30 patients with co-reactivation of CMV and EBV (CMV++EBV+ group), 101 patients with CMV viremia (CMV+ group), 149 patients with EBV viremia (EBV+ group), and 28 patients with CMV and EBV inactivation (CMV-+ EBV- group). Compared with the other groups, the incidence of acute graft-versus-host disease (aGVHD) and hemorrhagic cystitis (HC) was higher in CMV++ EBV+ groups (53.3% vs 42.6%, 36.9%, 17.9%, P < 0.001; 36.7% vs 32.7%, 22.8%, 10.7%, P =0.042). The incidence of post-transplant lymphoproliferative disease (PTLD) in CMV++ EBV+ group was similar to CMV+ group and EBV+ group (3.3% vs 3.0%, 3.4%, P =0.811). Univariate and multivariate analysis showed that the persistent time of CMV and EBV after transplantation were independent risk factors for co-reactivation of CMV and EBV. Compared with the other groups, the 2-year overall survival (OS) rate and 2-year disease-free survival (DFS) rate of patients in CMV++EBV+ group were lower (46.7% vs 74.9%, 83.4%, 71.4%, P < 0.001; 46.7% vs 70.9%, 79.5%, 69.9%, P =0.002), and 2-year non-recurrence mortality (NRM) was higher (48.2% vs 22%, 13.6%, 18.7%, P <0.001). CONCLUSION: The persistent time of CMV and EBV after transplantation are independent risk factors for patients with co-reactivation of CMV and EBV. Patients with co-reactivation of CMV and EBV had lower OS and DFS rate and higher NRM, suggesting that the clinical prognosis of the patients are worse.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Infecções por Vírus Epstein-Barr , Transplante de Células-Tronco Hematopoéticas , Herpesvirus Humano 4 , Transplante Homólogo , Humanos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Fatores de Risco , Doença Enxerto-Hospedeiro , Prognóstico , Ativação Viral , Feminino , Masculino , Viremia
18.
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
19.
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
20.
Pflugers Arch ; 465(10): 1439-49, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23686296

RESUMO

Transient receptor potential (TRP) channels are not well understood in human atrium, and the present study was therefore designed to investigate whether TRPC channels would mediate the nonselective cation current reported previously and are involved in the formation of store-operated Ca(2+) entry (SOCE) channels in human atrial myocytes using approaches of whole-cell patch voltage-clamp, RT-PCR, Western blotting, co-immunoprecipitation, and confocal scanning approaches, etc. We found that a nonselective cation current was recorded under K(+)-free conditions in human atrial myocytes, and the current was inhibited by the TRP channel blocker La(3+). Thapsigargin enhanced the current, and its effect was suppressed by La(3+) and prevented by pipette inclusion of anti-TRPC1 antibody. Endothlin-1 and angiotensin II enhanced the current that could be inhibited by La(3+). Gene and protein expression of TRPC1 channels were abundant in human atria. In addition, mRNA and protein of STIM1 and Orai1, components of SOCE channels, were abundantly expressed in human atria. Co-immunoprecipitation analysis demonstrated an interaction of TRPC1 with STIM1 and/or Orai1. Ca(2+) signaling mediated by SOCE channels was detected by a confocal microscopy technique. These results demonstrate the novel evidence that TRPC1 channels not only mediate the nonselective cation current, but also form SOCE channels in human atria as a component. TRPC1 channels can be activated by endothelin-1 or angiotensin II, which may be involved in the atrial electrical remodeling in patients with atrial fibrillation.


Assuntos
Átrios do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Cátion TRPC/metabolismo , Potenciais de Ação , Angiotensina II/farmacologia , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Células Cultivadas , Endotelina-1/farmacologia , Átrios do Coração/citologia , Humanos , Lantânio/farmacologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Tapsigargina/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa