Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Intern Med J ; 51(5): 732-738, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32149434

RESUMO

BACKGROUND: Primary insomnia is a worldwide problem and it has a considerable negative impact on one's physical and mental health. Studies have shown that non-synonymous Single-nucleotide polymorphisms in 5-hydroxytryptamine (serotonin or 5-HT) are related to primary insomnia. Previous studies have shown that 5-HT polymorphism (rs140700) is related to depression, and insomnia is often accompanied by depression and anxiety. The relationship between this site and primary insomnia is unknown. We speculated that this site may be related to primary insomnia, so we investigated the relationship between rs140700 and primary insomnia. AIMS: To explore the relationship between the 5-HT gene polymorphism rs140700 and primary insomnia. METHODS: In this study, we included 57 patients with primary insomnia and 54 age- and gender-matched normal controls. The subjects who belonged to the Chinese population were subjected to polysomnography for three consecutive nights. Their sleep quality was assessed, and the genotypes of the 5-hydroxytryptamine (5-HT) gene polymorphism rs140700 were determined by the flight mass spectrometry. RESULTS: The genotype distributions of the 5-HT gene polymorphism rs140700 were in Hardy-Weinberg equilibrium in both patients and controls (P > 0.05). The allele and genotype distributions of this variant were comparable between the patients and controls in all subjects and between genders (all P > 0.05). The influence of rs140700 on percentage of stage 1 (P = 0.015) change and arousal index (P = 0.028) of primary insomnia was statistically significant. The logistic multi-factor regression analysis results revealed that 5-HT gene polymorphism rs140700 was not a risk factor for primary insomnia in the Chinese population (P = 0.589). CONCLUSIONS: The 5-HT gene polymorphism rs140700 may not be a susceptibility locus for primary insomnia in the Chinese population.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Serotonina , Distúrbios do Início e da Manutenção do Sono , Alelos , Povo Asiático/genética , Estudos de Casos e Controles , China/epidemiologia , Feminino , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , Distúrbios do Início e da Manutenção do Sono/epidemiologia , Distúrbios do Início e da Manutenção do Sono/genética
2.
Biochim Biophys Acta ; 1863(7 Pt A): 1579-88, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27066979

RESUMO

Resident cardiac Sca-1-positive (+) stem cells may differentiate into cardiomyocytes to improve the function of damaged hearts. However, little is known about the inducers and molecular mechanisms underlying the myogenic conversion of Sca-1(+) stem cells. Here we report that sphingosylphosphorylcholine (SPC), a naturally occurring bioactive lipid, induces the myogenic conversion of Sca-1(+) stem cells, as evidenced by the increased expression of cardiac transcription factors (Nkx2.5 and GATA4), structural proteins (cardiac Troponin T), transcriptional enhancer (Mef2c) and GATA4 nucleus translocation. First, SPC activated JNK and STAT3, and the JNK inhibitor SP600125 or STAT3 inhibitor stattic impaired the SPC-induced expression of cardiac transcription factors and GATA4 nucleus translocation, which suggests that JNK and STAT3 participated in SPC-promoted cardiac differentiation. Moreover, STAT3 activation was inhibited by SP600125, whereas JNK was inhibited by ß-cyclodextrin as a lipid raft breaker, which indicates a lipid raft/JNK/STAT3 pathway involved in SPC-induced myogenic transition. ß-Catenin, degraded by activated GSK3ß, was inhibited by SPC. Furthermore, GSK3ß inhibitors weakened but the ß-catenin inhibitor promoted SPC-induced differentiation. We found no crosstalk between the lipid raft/JNK/STAT3 and ß-catenin pathway. Our study describes a lipid, SPC, as an endogenic inducer of myogenic conversion in Sca-1(+) stem cells with low toxicity and high efficiency for uptake.


Assuntos
Antígenos Ly/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Fosforilcolina/análogos & derivados , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Células-Tronco/efeitos dos fármacos , beta Catenina/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Microdomínios da Membrana/enzimologia , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/enzimologia , Fosforilcolina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Esfingosina/farmacologia , Células-Tronco/enzimologia , Fatores de Tempo , beta Catenina/antagonistas & inibidores
3.
Apoptosis ; 21(5): 546-57, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26921179

RESUMO

Elevated circulatory free fatty acids (FFAs) especially saturated FFAs, such as palmitate (PA), are detrimental to the heart. However, mechanisms responsible for this phenomenon remain unknown. Here, the role of JAK2/STAT3 in PA-induced cytotoxicity was investigated in cardiomyocytes. We demonstrate that PA suppressed the JAK2/STAT3 pathway by dephosphorylation of JAK2 (Y1007/1008) and STAT3 (Y705), and thus blocked the translocation of STAT3 into the nucleus. Conversely, phosphorylation of S727, another phosphorylated site of STAT3, was increased in response to PA treatment. Pretreatment of JNK inhibitor, but not p38 MAPK inhibitor, inhibited STAT3 (S727) activation induced by PA and rescued the phosphorylation of STAT3 (Y705). The data suggested that JNK may be another upstream factor regulating STAT3, and verified the important function of P-STAT3 (Y705) in PA-induced cardiomyocyte apoptosis. Sodium orthovanadate (SOV), a protein tyrosine phosphatase inhibitor, obviously inhibited PA-induced apoptosis by restoring JAK2/STAT3 pathways. This effect was diminished by STAT3 inhibitor Stattic. Collectively, our data suggested a novel mechanism that the inhibition of JAK2/STAT3 activation was responsible for palmitic lipotoxicity and SOV may act as a potential therapeutic agent by targeting JAK2/STAT3 in lipotoxic cardiomyopathy treatment.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Janus Quinase 2/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Palmitatos/toxicidade , Fator de Transcrição STAT3/metabolismo , Vanadatos/farmacologia , Animais , Linhagem Celular , Células Cultivadas , Janus Quinase 2/antagonistas & inibidores , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Palmitatos/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Ratos Sprague-Dawley , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
4.
J Cardiovasc Pharmacol ; 67(4): 312-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26751265

RESUMO

Vascular endothelial cell (VEC) apoptosis is involved in the development of atherosclerosis and other cardiovascular diseases. We previously found that ethyl 1-(2-hydroxy-3-aroxypropyl)-3-aryl-1H-pyrazole -5-carboxylate derivatives (3a-o) play important roles in cell fate control. In this study, among the 15 compounds, we further screened 2 compounds, 3d and 3k, that suppressed VEC apoptosis induced by deprivation of serum and fibroblast growth factor 2. To clarify which chiral enantiomers of 3d and 3k functioned, we synthesized 3d-S and its enantiomer 3d-R, 3k-S, and its enantiomer 3k-R. Then, we investigated the apoptosis-inhibiting activity of the chiral compounds in VECs. Four small molecules, 3d-S, 3d-R, 3k-S, 3k-R, significantly elevated VEC viability and inhibited apoptosis. Furthermore, these small molecules could obviously decrease the level of integrin ß4 that plays a key role in the regulation of VEC apoptosis. 3k-S and 3k-R increased Bcl-2/Bax ratio and reduced reactive oxygen species levels dramatically. Therefore, we provide new VEC apoptosis inhibitors. These compounds may be potential agents in the prevention of vascular diseases associated with VEC apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Pirazóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Integrina beta4/metabolismo , Pirazóis/química , Estereoisomerismo , Doenças Vasculares/prevenção & controle
5.
Anal Chem ; 87(24): 12088-95, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26585577

RESUMO

Herein, we reported a red-emitting probe (E)-4-(2-(8-hydroxy-julolidine-9-yl)vinyl)-1-methylpyridin-1-ium iodide (HJVPI) on a rotor mechanism with an ultrahigh signal-to-noise ratio. HJVPI could give high-fidelity fluorescent images of mitochondria in living immortalized and normal cells and be suitable for IR excitation source of two-photon microscopy and various excitation sources of confocal microscopy. As a rotor, its single/two-photon fluorescence intensities directly depended on environmental viscosity. And, as a mitochondrial probe, it displayed much larger two-photon absorption cross sections in comparison with commercial MitoTracker Green FM and MitoTracker Red FM. Moreover, the fact that living cells stained by HJVPI still possessed physiological function could also be confirmed: (1) MTT assay demonstrated that the mitochondria of cells stained retained their electron mediating ability and (2) double assay of HJVPI and SYTOX Blue nucleic acid stain (S-11348) showed that the plasma membrane of the cells stained was still intact. In addition, HJVPI possessed a number of beneficial properties in bioimaging such as good membrane permeability, high photostability, and excellent counterstain compatibility with Hoechst 33342. Related mechanism research suggested that its localization property was dependent on the mitochondrial membrane potential in living cells. All its remarkable properties can extend the investigation on mitochondria in a biological context.


Assuntos
Técnicas Citológicas/instrumentação , Mitocôndrias , Sobrevivência Celular , Citometria de Fluxo , Células HeLa , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Razão Sinal-Ruído , Coloração e Rotulagem
6.
Biochem Biophys Res Commun ; 463(3): 262-7, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26002468

RESUMO

Palmitate (PA), one of the most prevalent saturated fatty acids, causes myocardial dysfunction. However, the mechanisms by which PA induces cell apoptosis and autophagy remain to be elucidated. We showed that autophagy was induced in an mTORC1-dependent way and played a protective role against PA-induced apoptosis, which was verified by pretreatment with 3-methyladenine (3MA) and rapamycin. However, p62 began to accumulate after 18 h treatment with PA, suggesting prolonged exposure to PA lead to an impairment of autophagic flux. PA enhanced ROS production as well as activated p38-mitogen-activated protein kinase (p38 MAPK) and c-jun NH2 terminal kinases (JNKs). The antioxidant N-Acety-l-Cysteine (NAC) was found to attenuate the JNK and p38 MAPK activation with a concomitant reduction of PA-induced autophagy and apoptosis. Furthermore, both JNK and p38 MAPK inhibitors were shown to directly abrogate caspase 7 cleavage as well as the conversion of LC3BI to LC3BII. Thus, we demonstrate that PA stimulates autophagy and apoptosis via ROS-dependent JNK and p38 MAPK pathways.


Assuntos
Autofagia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Palmitatos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , Miocárdio/citologia , Ratos , Serina-Treonina Quinases TOR/metabolismo
7.
Fish Shellfish Immunol ; 47(1): 271-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26363234

RESUMO

Toll-like receptors (TLRs) are important pattern recognition receptors in the innate immune system of fish. Although ten years have passed since the first identification, the systematic knowledge about fish-specific TLR19 is still far insufficient. In present study, a phylogenetic analysis showed that TLR19 belonged to family 11, and clustered with TLR20 and TLR11/12 on the evolutionary tree. TLR20 is the closest paralogue of TLR19. The ectodomain of TLR19 contains 24 leucine-rich repeat (LRR) modules. The electrostatic surface potential analysis indicated that the modeled structure of TLR19 ectodomain showed much stronger polarity on the ascending lateral surface than on the descending lateral surface. The ascending lateral surface with strong electrostatic surface potential possibly mainly participates in the ligand binding of TLR19 ectodomain. The quite small dN/dS value at the TLR19 locus showed that TLR19 was very conserved. Approximately one third codons in the coding sequence of TLR19 were subjected to significantly negative selection, whereas only 5 codons underwent significantly positive selection. Overall, these findings possibly help in deepening the understanding to fish-specific TLR19.


Assuntos
Evolução Molecular , Proteínas de Peixes/genética , Peixes/genética , Receptores Toll-Like/genética , Sequência de Aminoácidos , Animais , Proteínas de Peixes/química , Proteínas de Peixes/metabolismo , Peixes/imunologia , Peixes/metabolismo , Conformação Molecular , Filogenia , Receptores Toll-Like/química , Receptores Toll-Like/metabolismo
8.
Cell Biol Toxicol ; 31(1): 15-27, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25575676

RESUMO

Nano-Mg(OH)2 is efficiently used in pollutant adsorption and removal due to its high adsorption capability, low-cost, and recyclability. A recent research from our group showed that Mg(OH)2 nanoflakes are not evidently internalized by cancer cells and are not cytotoxic. But the biocompatibility and potential toxicity of nano-Mg(OH)2 in a normal biological system are largely unclear. Nanoparticles could affect the function of endothelial cells, and endothelial dysfunction represents an early sign of lesion within the vasculature. Here, we applied the human umbilical vein vascular endothelial cells (HUVECs) as an in vitro model of the endothelium to study the cytotoxicity of nano-Mg(OH)2. Our results showed that nano-Mg(OH)2 at 200 µg/ml impaired proliferation and induced dysfunction of HUVECs, but did not result in cell necrosis and apoptosis. Transmission electron microscopy images and immunofluorescence results showed that the nano-Mg(OH)2 could enter HUVECs through caveolin-1-mediated endocytosis. Nano-Mg(OH)2 at high concentrations decreased the level of caveolin-1 and increased the activity of endothelial nitric oxide synthase (eNOS), thus leading to the production of excess nitric oxide (NO). In this work, we provide the cell damage concentrations of nano-Mg(OH)2 nanoparticles, and we propose a mechanism of injury induced by nano-Mg(OH)2 in HUVECs.


Assuntos
Caveolina 1/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Hidróxido de Magnésio/toxicidade , Nanopartículas Metálicas/toxicidade , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Endocitose , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Hidróxido de Magnésio/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo
9.
Environ Toxicol ; 30(7): 755-68, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24449189

RESUMO

ZnS nanoarchitectures have been intensively investigated recently because of their applications in optoelectronics and adsorption capacity. The potential hazard of ZnS nanoarchitectures is not well known. In this study, we investigated the toxicity of ZnS nanoarchitectures on vascular endothelial cell (VEC) in vitro and in vivo. The results showed that ZnS could inhibit human umbilical vein endothelial cell (HUVEC) proliferation at 50 and 200 µg/mL. Endothelial nitric oxide synthase (eNOS) activity, nitric oxide (NO), and reactive oxygen species productions were increased, which was companied with the decrease in caveolin-1 level. The endothelium of the aortic root was damaged and the NO levels in serum were elevated in the mice treated with 5 or 10 mg/kg ZnS for 3 and 6 days, but the body could repair the damage. The data suggested that the high concentration of ZnS could induce dysfunction of VECs through decreasing caveolin-1 and elevation of the eNOS activity and thus present toxicity.


Assuntos
Proliferação de Células/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Sulfetos/química , Compostos de Zinco/química , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Apoptose/efeitos dos fármacos , Caveolina 1/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Imuno-Histoquímica , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Óxido Nítrico/sangue , Óxido Nítrico Sintase Tipo III/metabolismo , Espécies Reativas de Oxigênio/metabolismo
10.
Biochim Biophys Acta ; 1833(9): 2092-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23651924

RESUMO

Autophagy involves multiple membrane trafficking and fusion events. Annexin A7 (ANXA7) is postulated to play a role in membrane fusion during exocytosis, while the contribution of ANXA7 to autophagy is poorly understood. Our recent studies demonstrated that ABO could promote autophagy via elevation of ANXA7 and triggering ANXA7 subcellular redistribution. However, little is known about the molecular mechanisms how ANXA7 regulates autophagy. As molecular disruption of ANXA7 in mice results in several unwished phenotypes, small molecule modulators may be efficacious in defining the mechanisms of ANXA7 action. However, so far no compounds that selectively target ANXA7 have been identified. So, we hypothesize that ABO might be a potent modulator of ANXA7. We also have detected the colocalization of ANXA7 and microtubule-associated protein 1 light chain 3 (LC3), and ANXA7 was essential for LC3 accumulation in VEC autophagy. As a GTPase, whether ANXA7 affects the phosphorylation of LC3 or other proteins needs further investigation. In this study, we performed site-directed mutagenesis and found that ABO directly bound to Thr(286) of ANXA7 and inhibited its phosphorylation. By yeast two-hybrid screening, we found that ANXA7 could interact with grancalcin (GCA). ABO promoted the interaction and inhibited GCA phosphorylation, leading to the decrease of intracellular Ca(2+) concentration. At the same time, ABO inhibited the phosphorylation of LC3. Hence, by identifying ABO as an unprecedented modulator of ANXA7 as well as GCA and LC3 as interacting proteins of ANXA7, we demonstrated the possible mechanisms how ANXA7 regulates autophagy for the first time.


Assuntos
Anexina A7/antagonistas & inibidores , Anexina A7/metabolismo , Autofagia/efeitos dos fármacos , Benzoxazinas/farmacologia , Animais , Anexina A7/genética , Benzoxazinas/química , Células COS , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Chlorocebus aethiops , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação/efeitos dos fármacos
11.
J Physiol ; 591(20): 5005-15, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23959677

RESUMO

We previously found that phosphatidylcholine-specific phospholipase C (PC-PLC) was a key inducing element of atherosclerosis, and might negatively regulate human umbilical vein endothelial cell (HUVEC) autophagy. To further investigate the mechanism of PC-PLC action, we initially identified phosphatidylethanolamine binding protein 1 (PEBP1) as a binding partner of PC-PLC by using mass spectrometry (MS, MALDI-TOF/TOF). We found that PEBP1 positively regulated PC-PLC activity in HUVECs, and inhibition of PC-PLC by its inhibitor D609 suppressed PEBP1 expression dramatically. Moreover, both PC-PLC and PEBP1 negatively regulated HUVEC autophagy independently of mammalian target of rapamycin (mTOR). Furthermore, the PEBP1 level was elevated during the development of atherosclerosis, while D609 significantly decreased the upregulated PEBP1 level in apoE(-/-) mice.


Assuntos
Aterosclerose/metabolismo , Autofagia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Animais , Apolipoproteínas E/genética , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Norbornanos , Proteína de Ligação a Fosfatidiletanolamina/genética , Ligação Proteica , Serina-Treonina Quinases TOR/metabolismo , Tiocarbamatos , Tionas/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
12.
Apoptosis ; 18(9): 1120-31, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23677256

RESUMO

Integrin ß4 and its Y-1494 phosphorylation play an important role in cell signaling. We found a small molecule, ethyl1-(3-(4-chlorophenoxy)-2-hydroxypropyl)-3-(4-chlorophenyl)-1H-pyrazole-5-carboxylate (ECPC), that could elevate the levels of KIT ligand (KITLG), interleukin 8 (IL-8), prostaglandin-endoperoxide synthase 2 (PTGS2) and activating transcription factor 3 (ATF3) and promote apoptosis in vascular endothelial cells (VECs) through integrin ß4. We investigated the underlying mechanism of integrin ß4 participating in this process. ECPC treatment increased the phosphorylation of Y-1494 in the integrin ß4 cytoplasmic domain via a well-known receptor tyrosine kinase, fibroblast growth factor receptor 1 (FGFR1), and integrin ß4 translocated from the cytoplasm to nucleus. With suppression of Y-1494 phosphorylation by FGF-2 or siRNA of FGFR1, ECPC failed to promote integrin ß4 nuclear translocation and could not increase the expression of KITLG, IL-8, PTGS2 or ATF3. Y-1494 phosphorylation and nuclear translocation of integrin ß4 may be important during ECPC-induced apoptosis in VECs.


Assuntos
Apoptose , Núcleo Celular/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Integrina beta4/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Motivos de Aminoácidos , Apoptose/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Integrina beta4/química , Integrina beta4/genética , Fosforilação/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Bibliotecas de Moléculas Pequenas/síntese química
13.
Acta Pharmacol Sin ; 34(7): 960-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23645009

RESUMO

AIM: To investigate the effects of 7 novel 1-ferrocenyl-2-(5-phenyl-1H-1,2,4-triazol-3-ylthio) ethanone derivatives on human lung cancer cells in vitro and to determine the mechanisms of action. METHODS: A549 human lung cancer cells were examined. Cell viability was analyzed with MTT assay. Cell apoptosis and senescence were examined using Hoechst 33258 and senescence-associated-ß-galactosidase (SA-ß-gal) staining, respectively. LDH release was measured using a detection kit. Cell cycle was analyzed using a flow cytometer. Intracellular ROS level was measured with the 2',7'-dichlorodihydrofluorescein probe. Phosphorylation of p38 was determined using Western blot. RESULTS: Compounds 5b, 5d, and 5e (40 and 80 µmol/L) caused significant decrease of A549 cell viability, while other 4 compounds had no effect on the cells. Compounds 5b, 5d, and 5e (80 µmol/L) induced G1-phase arrest (increased the G1 population by 22.6%, 24.23%, and 26.53%, respectively), and markedly increased SA-ß-gal-positive cells. However, the compounds did not cause nuclear DNA fragmentation and chromatin condensation in A549 cells. Nor did they affect the release of LDH from the cells. The compounds significantly elevated the intracellular ROS level, decreased the mitochondrial membrane potential, and increased p38 phosphorylation in the cells. In the presence of the antioxidant and free radical scavenger N-acetyl-L-cysteine (10 mmol/L), above effects of compounds 5b, 5d, and 5e were abolished. CONCLUSION: The compounds 5b, 5d, and 5e cause neither apoptosis nor necrosis of A549 cells, but exert anti-cancer effect via inducing G1-phase arrest and senescence through ROS/p38 MAP-kinase pathway.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Senescência Celular/efeitos dos fármacos , Compostos Ferrosos/química , Compostos Ferrosos/farmacologia , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Neoplasias Pulmonares , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Senescência Celular/fisiologia , Compostos Ferrosos/uso terapêutico , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Metalocenos , Espécies Reativas de Oxigênio/metabolismo
14.
Acta Pharmacol Sin ; 33(1): 57-65, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22139003

RESUMO

AIM: In vascular strips, the adjacent endothelial cells modulate the contraction of vascular smooth muscle cells (VSMCs) induced by sphingosylphosphorylcholine (SPC) through nitric oxide (NO). The aim of this study was to elucidate the mechanisms by which vascular endothelial cells (VECs) reduce the SPC-induced contraction of VSMCs in a co-culture system. METHODS: Human umbilical VECs and VSMCs were co-cultured. The VECs were transfected with integrin ß4- or Fyn-specific siRNA. The areas of VSMCs that are involved in cell contractility were quantified using the Leica confocal software and collagen contractility assay. The production of NO in VECs was measured in the cell supernatants using NO Detection Kit. The levels of integrin ß4 and Fyn in VECs and the levels of Rho kinase (ROCK) in VSMC were detected using immunofluorescence assays or Western blots. RESULTS: Co-culture with VECs reduced the contraction of VSMCs induced by SPC (30 µmol/L). The down-regulation of integrin ß4 or Fyn in VECs by the specific siRNA (20 nmol/L) was able to counteract the effects of VECs on the SPC-induced VSMC contractions. Furthermore, the integrin ß4-specific siRNA (20 and 40 nmol/L) significantly reduced the level of Fyn protein and the production of NO in VECs, while increased the level of ROCK in VSMCs that had been stimulated by SPC. CONCLUSION: The VECs reduced the SPC-induced contraction of VSMCs in the co-culture system through integrin ß4 and Fyn proteins. In this process, NO may be the factor downstream of integrin ß4 in VECs, while ROCK may be the key protein regulating the contraction of VSMCs.


Assuntos
Células Endoteliais/fisiologia , Integrina beta4/metabolismo , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fosforilcolina/análogos & derivados , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Esfingosina/análogos & derivados , Animais , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/citologia , Humanos , Integrina beta4/genética , Contração Muscular/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/fisiologia , Óxido Nítrico/metabolismo , Fosforilcolina/farmacologia , Proteínas Proto-Oncogênicas c-fyn/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Esfingosina/farmacologia , Quinases Associadas a rho/metabolismo
15.
Cells ; 11(19)2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36231088

RESUMO

Human dermal fibroblasts (HDFs) have the potential to differentiate into endothelial cells (VECs). In our previous research, we reported that a hypochlorous acid (HOCl) probe CPP efficiently induced the differentiation of HDFs into VECs, however, the mechanism of differentiation was not clear. As an HOCI probe, CPP binds HOCI to modulate its effects. In this study, through Western blotting, qPCR, and PHD2 enzyme activity assay, we found that CPP inhibited the enzyme activity of prolyl-4-hydroxylase 2 (PHD2), thereby stabilizing HIF-1α. To further clarify the mechanism by which CPP inhibits PHD2 enzyme activity, we constructed plasmids, and found that CPP inhibited PHD2 activity to increase the HIF-1α level through the modulation of PHD2 at Cys302 by HOCl in HDFs. Furthermore, RNA-seq experiments showed that CPP could induce the expression of HEY1, which is not only a target gene regulated by HIF1α, but also a key transcription factor for VECs. We used siRNA transfection and in vivo experiments to confirm that CPP could induce HDFs to differentiate into VECs by HEY1. In summary, we identified a new inhibitor of PHD2, demonstrated the new role of HOCl in cell differentiation, and elucidated the mechanism by which HOCl probe CPP induced the differentiation of HDFs into VECs.


Assuntos
Células Endoteliais , Prolina Dioxigenases do Fator Induzível por Hipóxia , Humanos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Ácido Hipocloroso/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolil Hidroxilases/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais
16.
Genes (Basel) ; 13(9)2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36140818

RESUMO

Researchers are paying more and more attention to aging, especially skin aging. Therefore, it is urgent to find an effective way to inhibit aging. Here, we report a small chemical molecule, HCP1, that inhibited the senescence of human dermal fibroblasts (HDFs). First, we performed morphological experiment and found that HCP1-treated HDFs were no longer elongated and flat compared to DMSO-treated groups. Next, we found that the number of ß-gal positive cells decreased compared to DMSO-treated groups. Through flow cytometry, western blot, and immunofluorescence, we found that HCP1 could inhibit the senescence of HDFs. In the study of the mechanism, we found that HCP1 could regulate the AMPK/mTOR signal pathway through glucose-regulated protein 94 (Grp94). In addition, we found that HCP1 could promote the interaction between Grp94 and lysosomes, which led to an increase in the activity of lysosomes and inhibited the senescence of HDFs. At the same time, we found that HCP1 decreased the concentration of Ca2+ in mitochondria, inhibiting the senescence of HCP1. Therefore, we propose that HCP1 is a potential aging-inhibiting compound, and provide a new idea for the development of senescence-inhibiting drugs.


Assuntos
Proteínas Quinases Ativadas por AMP , Senescência Celular , Proteínas Quinases Ativadas por AMP/metabolismo , Dimetil Sulfóxido/farmacologia , Fibroblastos/metabolismo , Proteínas de Choque Térmico HSP70 , Humanos , Proteínas de Membrana , Serina-Treonina Quinases TOR/metabolismo
17.
J Cell Physiol ; 226(11): 2827-33, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21302284

RESUMO

Sphingosylphosphorylcholine (SPC), an important cardiovascular mediator derived from sphingomyelin that has atheroprotective effects via actions on vascular endothelial cells (VECs) at normal levels in vivo. However, the underlying mechanism is not well known. To clarify this question, we first investigated the effect of SPC on VEC apoptosis and autophagy induced by deprivation of serum and fibroblast growth factor 2 (FGF-2). SPC at 5-20 µM inhibited apoptosis and induced autophagy in vitro. To understand the underlying mechanism, we investigated the role of integrin ß4 in SPC-induced autophagy in VECs. SPC significantly decreased the level of integrin ß4, whereas overexpression of integrin ß4 inhibited SPC-induced autophagy. Moreover, knockdown of integrin ß4 promoted VEC autophagy. To understand the downstream factors of integrin ß4 in this process, we observed the effects of SPC on phosphatidylcholine-specific phospholipase C (PC-PLC) activity and level of p53. PC-PLC activity and p53 level in cytoplasm was decreased during autophagy induced by SPC, and knockdown of integrin ß4 inhibited the activity of PC-PLC and the cytoplasmic level of p53. SPC may promote autophagy via integrin ß4. Moreover, PC-PLC and p53 may be the downstream factors of integrin ß4 in autophagy of VECs deprived of serum and FGF-2.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Fosforilcolina/análogos & derivados , Esfingosina/análogos & derivados , Células Cultivadas , Células Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Integrina beta4/metabolismo , Fosforilcolina/farmacologia , Esfingosina/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Fosfolipases Tipo C/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 30(3): 411-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20139365

RESUMO

OBJECTIVE: Atherosclerosis is considered to be a chronic inflammatory disease. Previous research has demonstrated that phosphatidylcholine-specific phospholipase C (PC-PLC) plays critical roles in various inflammatory responses. However, the association between PC-PLC and atherosclerosis is undetermined. Therefore, we sought to investigate whether PC-PLC was implicated in atherosclerosis. METHODS AND RESULTS: Immunofluorescence analysis revealed an upregulation of PC-PLC in the aortic endothelium from apolipoprotein E-deficient (apoE(-/-)) mice. PC-PLC level and activity were also increased in human umbilical vein endothelial cells in response to oxidized low-density lipoprotein treatment. Pharmacological blockade of PC-PLC by D609 inhibited the progression of preexisting atherosclerotic lesions in apoE(-/-) mice and changed the lesion composition into a more stable phenotype. Using a combination of pharmacological inhibition, polyclonal antibodies, confocal laser scanning microscopy and Western blotting, we demonstrated that PC-PLC was required for endothelial expression of lectin-like oxidized low-density lipoprotein receptor-1. In addition, D609 treatment significantly decreased the aortic endothelial expression of the vascular cell adhesion molecule-1 and the intercellular adhesion molecule-1. Furthermore, inhibition of PC-PLC in human umbilical vein endothelial cells reduced the oxidized low-density lipoprotein induced expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and monocyte chemotactic protein-1. CONCLUSIONS: Our data suggest that PC-PLC contributes to the progression of atherosclerosis.


Assuntos
Apolipoproteínas E/fisiologia , Aterosclerose/tratamento farmacológico , Aterosclerose/fisiopatologia , Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Progressão da Doença , Tionas/uso terapêutico , Fosfolipases Tipo C/fisiologia , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Knockout , Norbornanos , Tiocarbamatos , Tionas/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Regulação para Cima/fisiologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
19.
Acta Pharmacol Sin ; 32(2): 209-16, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21293473

RESUMO

AIM: To investigate the effect of N-benzyl-5-phenyl-1H-pyrazole-3-carboxamide (BPC) on angiogenesis in human umbilical vein endothelial cells (HUVECs). METHODS: Capillary-like tube formation on matrigel and cell migration analyses were performed in the absence of serum and fibroblast growth factor (FGF-2). Reactive oxygen species (ROS) were measured using a fluorescent probe, 2', 7'- dichlorodihydrofluorescein (DCHF). The nitric oxide (NO) production of HUVECs was examined using a NO detection kit. Morphological observation under a phase contrast microscope, a viability assay using 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl-tetrazolium (MTT) and a lactate dehydrogenase (LDH) activity analysis by a detection kit were performed to evaluate the toxicity of BPC on HUVECs in the presence of serum and FGF-2. The level of hypoxia-inducible factor 1α (HIF-1α) and the release of vascular endothelial growth factor (VEGF) were measured by Western blot and ELISA, respectively. RESULTS: In the absence of serum and FGF-2, cells treated with BPC (5-20 µmol/L) rapidly aligned with one another and formed tube-like structures within 12 h. In the presence of serum and FGF-2, cells treated with BPC for 24, 48 and 72 h had no changes in morphology, viability or LDH release compared with the control group. Cell migration in the BPC-treated group was significantly increased compared with the control group. During this process, NO production and ROS level were elevated dramatically, and the levels of HIF-1α and VEGF were increased dependent on the generation of ROS. CONCLUSION: BPC most effectively promoted angiogenesis and migration in HUVECs in the absence of FGF-2 and serum.


Assuntos
Compostos de Benzil/farmacologia , Células Endoteliais/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Pirazóis/farmacologia , Compostos de Benzil/administração & dosagem , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , L-Lactato Desidrogenase/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Pirazóis/administração & dosagem , Espécies Reativas de Oxigênio , Fatores de Tempo , Veias Umbilicais/citologia , Veias Umbilicais/efeitos dos fármacos , Veias Umbilicais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
J Cell Physiol ; 225(3): 673-81, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20509141

RESUMO

Increasing evidence has demonstrated that the senescence of vascular endothelial cells (VECs) has critical roles in the pathogenesis of vascular dysfunction. Finding important factors that regulate VEC senescence will help provide novel therapeutic strategies for vascular disorders. Previously, we found that integrin ß4 was involved in VEC senescence. However, the mechanism underlying VEC senescence mediated by integrin ß4 remains poorly understand. In this study, we used a mouse in vivo model and showed that the level of integrin ß4 in the endothelium of mouse thoracic aorta was increased during natural aging and atherosclerosis. Furthermore, we found that H-ras, caveolin-1, and AP-1 were implicated in the senescent signal pathway mediated by integrin ß4 in human umbilical vein ECs (HUVECs). Knockdown of integrin ß4 could attenuate HUVEC senescent features, including increased interleukin-8 (IL-8) release and decreased endothelial nitric oxide synthase (eNOS) and NO levels and mitochondrial membrane potential in vitro. Our findings provide new clues illustrating the mechanism of VEC senescence. Integrin ß4 might be a potential target for therapy in cardiovascular diseases.


Assuntos
Senescência Celular , Células Endoteliais/metabolismo , Integrina beta4/metabolismo , Fatores Etários , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Caveolina 1/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Humanos , Integrina alfa6/metabolismo , Integrina beta4/genética , Interleucina-8/metabolismo , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Interferência de RNA , Fator de Transcrição AP-1/metabolismo , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA