Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res Perspect ; 7(3): e00487, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31149342

RESUMO

The phospholipid l-α-lysophosphatidylinositol (LPI), an endogenous ligand for GPR55, is elevated in patients with acute coronary syndrome, and a GPR55 antagonist cannabidiol (CBD) reduces experimental ischemia/reperfusion (I/R) injury. While LPI activates multiple signaling pathways, little is known about which ones are important in cardiomyocytes. In this study we explored whether activation of the Rho kinase/ROCK/p38 MAPK pathway is responsible for LPI-induced extension of I/R injury. Using a high-throughput screening method (dynamic mass redistribution; DMR), mouse- and human-induced pluripotent stem cell (iPSC) cardiomyocytes exposed to LPI were shown to exhibit a rapid, sustained, and concentration-dependent (1 nmol L-1-30 µmol L-1) cellular response. Y-27632 (ROCK inhibitor; 10 & 50 µmol L-1) and CBD (1 µmol L-1) both abolished the DMR response to LPI (10 µmol L-1). In murine iPSC cardiomyocytes, LPI-induced ROCK and p38 MAPK phosphorylation, both of which were prevented by Y-27632 and CBD, but did not induce JNK activation or cleavage of caspase-3. In hearts isolated from wild type (WT) mice subjected to 30 minutes global I/R, LPI (10 µmol L-1) administered via the coronary circulation increased infarct size when applied prior to ischemia onset, but not when given at the time of reperfusion. The exacerbation of tissue injury by LPI was not seen in hearts from GPR55-/- mice or in the presence of Y-27632, confirming that injury is mediated via the GPR55/ROCK/p38 MAPK pathway. These findings suggest that raised levels of LPI in the vicinity of a developing infarct may worsen the outcome of AMI.


Assuntos
Lisofosfolipídeos/efeitos adversos , Traumatismo por Reperfusão Miocárdica/induzido quimicamente , Receptores de Canabinoides/metabolismo , Quinases Associadas a rho/metabolismo , Amidas/farmacologia , Animais , Modelos Animais de Doenças , Ensaios de Triagem em Larga Escala , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Traumatismo por Reperfusão Miocárdica/metabolismo , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , Receptores de Canabinoides/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Sci Rep ; 9(1): 2662, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30804442

RESUMO

Lysophosphatidic acid (LPA) is a blood-derived bioactive lipid with numerous biological activities exerted mainly through six defined G protein-coupled receptors (LPA1-LPA6). LPA was first identified as a vasoactive compound because it induced transient hypertension when injected intravenously in rodents. Here, we examined the molecular mechanism underlying the LPA-induced hypertensive response. The LPA-induced hypertensive response was significantly attenuated by pretreatment with a Rho kinase inhibitor, which blocks Gα12/13 signaling. Consistent with this, the response was weakened in KO mice of LPA4, a Gα12/13-coupling LPA receptor. KO mice of another Gα12/13-coupling LPA receptor, LPA6, also showed an attenuated LPA-induced hypertensive response. However, LPA6 KO mice also displayed attenuated pressor responses to an adrenergic agent and abnormal blood vessel formation. Using several LPA analogs with varied affinity for each LPA receptor, we found a good correlation between the hypertensive and LPA4 agonistic activities. Incubated mouse plasma, which contained abundant LPA, also induced a hypertensive response. Interestingly the response was completely abolished when the plasma was incubated in the presence of an ATX inhibitor. Together, these results indicate that circulating LPA produced by ATX contributes to the elevation of blood pressure through multiple LPA receptors, mainly LPA4.


Assuntos
Suscetibilidade a Doenças , Hipertensão/etiologia , Hipertensão/metabolismo , Lisofosfolipídeos/efeitos adversos , Animais , Biomarcadores , Modelos Animais de Doenças , Imunofluorescência , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Hipertensão/fisiopatologia , Camundongos , Transdução de Sinais , Quinases Associadas a rho/metabolismo
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(11): 1369-1377, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30591146

RESUMO

Extracellular lysophosphatidic acid (LPA) and the G-protein-coupled LPA receptors (LPAR) are involved in cell migration and invasion and found in the human endometrium. However, underlying mechanisms resulting in cellular invasion have been rarely investigated. We used stromal endometrial T-HESC, epithelial endometriotic 12Z, 49Z and Ishikawa cells. Interestingly, proliferation of T-HESC cells was strongly increased after LPA treatment, whereas the epithelial cell lines only showed a moderate increase. LPA increased invasion of 12Z and 49Z strongly and significantly. The LPAR inhibitor Ki16425 (LPAR1/3) attenuated significantly LPA-induced invasiveness of 12Z, which was confirmed by LPAR1 and LPAR3 siRNAs, showing that both LPA receptors contribute to invasiveness of 12Z cells. Investigation of cell invasion with an antibody-based protease array revealed mainly differences in cathepsins and especially cathepsin B between 12Z compared to the less invasive Ishikawa. Stimulation with LPA showed a time- and dose-dependent increased secretion of cathepsin B which was inhibited by the Gq inhibitor YM-254890 and Gi/o inhibitor pertussis toxin in the 12Z cells, again highlighting the importance of LPAR1/3. The activity of intracellular and secreted cathepsin B was significantly upregulated in LPA-treated samples. Inhibition of cathepsin B with the specific inhibitor CA074 significantly reduced LPA-increased invasion of 12Z. Our results reveal a novel role of LPA-mediated secretion of cathepsin B which stimulated invasion of endometriotic epithelial cells mainly via LPAR1 and LPAR3. These findings may deepen our understanding how endometriotic cells invade into ectopic sites, and provide new insights into the role of LPA and cathepsin B in cellular invasion.


Assuntos
Catepsina B/metabolismo , Endometriose/metabolismo , Lisofosfolipídeos/efeitos adversos , Regulação para Cima , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Humanos , Isoxazóis/farmacologia , Propionatos/farmacologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Fatores de Tempo
4.
Oncotarget ; 8(26): 43543-43554, 2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-28402936

RESUMO

Radiation-induced fibrosis is widely considered as a common but forsaken phenomenon that can lead to clinical sequela and possibly vital impairments. Lysophosphatidic acid is a bioactive lipid involved in fibrosis and probably in radiation-induced fibrosis as suggested in recent studies. Lysophosphatidic acid is also a well-described pro-oncogenic factor, involved in carcinogenesis processes (proliferation, survival, angiogenesis, invasion, migration). The present review highlights and summarizes the links between lysophosphatidic acid and radiation-induced fibrosis, lysophosphatidic acid and radioresistance, and proposes lysophosphatidic acid as a potential central actor of the radiotherapy therapeutic index. Besides, we hypothesize that following radiotherapy, the newly formed tumour micro-environment, with increased extracellular matrix and increased lysophosphatidic acid levels, is a favourable ground to metastasis development. Lysophosphatidic acid could therefore be an exciting therapeutic target, minimizing radio-toxicities and radio-resistance effects.


Assuntos
Transformação Celular Neoplásica/induzido quimicamente , Fibrose/etiologia , Lisofosfolipídeos/efeitos adversos , Animais , Biomarcadores , Fibrose/metabolismo , Fibrose/patologia , Humanos , Neoplasias/complicações , Neoplasias/etiologia , Neoplasias/patologia , Neoplasias/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Radiação Ionizante , Radioterapia/efeitos adversos , Radioterapia/métodos , Transdução de Sinais , Índice Terapêutico
5.
Clin Exp Med ; 16(1): 37-47, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25596714

RESUMO

Breast cancer generally shows poor prognosis because of its invasion and metastasis. Lysophosphatidic acid (LPA) induces and aggravates cancer invasion and metastasis by activating its downstream signal pathways. RhoA/ROCK/MMP signaling was found one of the LPA-induced pathways, which may be involved in invasion of breast cancer. Furthermore, we investigated whether this pathway was involved in curcumin's effect against LPA-induced invasion. LPA incubation was used to enhance invasion of MCF-7 breast cancer cells. RhoA expression was knocked-down by siRNA technique. MTT assay was used to evaluate the proliferation. Transwell assay was utilized to investigate the invasion ability of MCF-7 cells. Real-time PCR and Western blotting were used to assess the expressions of RhoA, ROCK1, ROCK2, MMP2 and MMP9 at both translational and transcriptional levels. The RhoA and ROCK activities were also evaluated. LPA incubation significantly boosted invasion rate of MCF-7. RhoA silencing by siRNA dramatically inhibited LPA-enhanced invasion. Concurrently, RhoA and ROCK activities and expression levels of RhoA, ROCK1, ROCK2, MMP2 and MMP9 were down-regulated by RhoA siRNA transfection. In order to avoid influence of cytotoxicity of curcumin, concentrations below 45 µmol/L were selected to further investigate the mechanism of curcumin's anti-invasion effect. Invasion of LPA-incubated MCF-7 cells was impaired by curcumin in a concentration-dependent manner. Concurrently, RhoA and ROCK activities and expression levels of RhoA, ROCK1, ROCK2, MMP2 and MMP9 were down-regulated by curcumin in a concentration-dependent manner. In conclusion, RhoA/ROCK/MMPs pathway activation is involved in LPA-induced invasion in MCF-7 cells; curcumin inhibited LPA-induced invasion in MCF-7 cells by attenuating RhoA/ROCK/MMPs pathway.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Curcumina/farmacologia , Lisofosfolipídeos/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Invasividade Neoplásica , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
6.
FEBS Lett ; 588(17): 2971-7, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-24911199

RESUMO

Polyunsaturated fatty acids (PUFAs) have important pharmacological effects on mammalian cells. Here, we show that carboxyl group-containing PUFAs inhibit lysophosphatidic acid (LPA)-induced focal adhesion formation, thereby inhibiting migration and adhesion. Carboxyl group-containing PUFAs inhibit LPA-induced calcium mobilization, whereas ethyl ester-group containing PUFAs have no effect. In addition, carboxyl group-containing PUFAs functionally inhibit LPA-dependent RhoA activation. Given these results, we suggest that PUFAs may inhibit LPA-induced calcium/RhoA signaling pathways leading to focal adhesion formation. Carboxyl group-containing PUFAs may have a functional role in this regulatory mechanism.


Assuntos
Movimento Celular/efeitos dos fármacos , Ácidos Graxos Insaturados/farmacologia , Lisofosfolipídeos/efeitos adversos , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Humanos , Transdução de Sinais/efeitos dos fármacos
7.
Thromb Haemost ; 107(2): 356-68, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22234341

RESUMO

Impaired endothelial recovery after the implantation of drug-eluting stents is a major concern because of the increased risk for late stent thrombosis. The disruption of the chemokine axis CXCL12/CXCR4 inhibits neointima formation by blocking the recruitment of smooth muscle progenitor cells. To directly compare a CXCR4-targeting treatment strategy with drugs that are currently used for stent coating, we studied the effects of the CXCR4 antagonist POL5551 and the drug sirolimus on neointima formation. Apolipoprotein E-deficient mice were treated with POL5551 or sirolimus continuously for 28 days after a carotid wire injury. POL5551 inhibited neointima formation by 63% (for a dosage of 2 mg/kg/day) and by 70% (for a dosage of 20 mg/kg/day). In comparison, sirolimus reduced the neointimal area by 69%. In contrast to treatment with POL5551 during the first three days after injury, injection of POL5551 (20 mg/kg) once per day for 28 days diminished neointimal hyperplasia by 53%. An analysis of the cellular composition of the neointima showed a reduction in the relative smooth muscle cell (SMC) and macrophage content in mice that had been treated with a high dose of POL5551. In contrast, the diminished SMC content after sirolimus treatment was associated with a neointimal enrichment of macrophages. Furthermore, endothelial recovery was impaired by sirolimus, but not by POL5551. Therefore, the inhibition of CXCR4 by POL5551 is equally effective in preventing neointima formation as sirolimus, but POL5551 might be more beneficial because treatment with it results in a more stable lesion phenotype and because it does not impair re-endothelialisation.


Assuntos
Angioplastia , Implante de Prótese Vascular , Endotélio Vascular/efeitos dos fármacos , Lisofosfolipídeos/administração & dosagem , Neointima/prevenção & controle , Complicações Pós-Operatórias/prevenção & controle , Proteínas/administração & dosagem , Animais , Apolipoproteínas E/genética , Artérias Carótidas/patologia , Artérias Carótidas/cirurgia , Movimento Celular/efeitos dos fármacos , Reestenose Coronária/etiologia , Reestenose Coronária/prevenção & controle , Modelos Animais de Doenças , Stents Farmacológicos/estatística & dados numéricos , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Lisofosfolipídeos/efeitos adversos , Lisofosfolipídeos/química , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Neointima/etiologia , Complicações Pós-Operatórias/etiologia , Proteínas/efeitos adversos , Receptores CXCR4/antagonistas & inibidores , Sirolimo/administração & dosagem , Sirolimo/efeitos adversos
8.
Clin Res Hepatol Gastroenterol ; 35(2): 89-97, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21809485

RESUMO

Pruritus is frequently reported by patients with cholestatic hepatobiliary diseases such as primary biliary cirrhosis, primary sclerosing cholangitis, intrahepatic cholestasis of pregnancy and hereditary cholestatic syndromes, but may accompany almost any other liver disease. Increased concentrations of bile salts, histamine, progesterone metabolites or endogenous opioids have been controversially discussed as potential pruritogens in cholestasis in the past. Most recently, novel insights unravelled lysophosphatidic acid (LPA), a potent neuronal activator, as a potential pruritogen in pruritus of cholestasis. Nevertheless, the pathogenesis of pruritus in cholestasis is still not clearly defined and current antipruritic treatment strategies provide relief only in a part of the affected patients. Based on recent experimental and clinical findings, this review outlines the actual insight in pathogenesis of pruritus in cholestasis and summarizes evidence-based and experimental therapeutic interventions for cholestatic patients suffering from itch.


Assuntos
Hepatopatias/complicações , Prurido/tratamento farmacológico , Prurido/fisiopatologia , Anticolesterolemiantes/administração & dosagem , Colagogos e Coleréticos/administração & dosagem , Colangite Esclerosante/complicações , Colestase Intra-Hepática/complicações , Quimioterapia Combinada , Medicina Baseada em Evidências , Feminino , Humanos , Cirrose Hepática Biliar/complicações , Lisofosfolipídeos/efeitos adversos , Gravidez , Complicações na Gravidez/tratamento farmacológico , Complicações na Gravidez/fisiopatologia , Prurido/etiologia , Resultado do Tratamento
9.
Zhonghua Xin Xue Guan Bing Za Zhi ; 39(4): 301-4, 2011 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-21624303

RESUMO

OBJECTIVE: To investigate the effect and potential mechanism of lysophosphatidic acid (LPA) and antiarrhythmic peptide (AAP10) on rabbit ventricular arrhythmia. METHODS: Twenty-four rabbits were randomly divided into three groups (n = 8 each): control group, LPA group and AAP10 + LPA group. Using arterially perfused rabbit ventricular wedge preparations, transmural ECG and action potentials from both endocardium and epicardium were simultaneously recorded in the whole process of all experiments with two separate floating microeletrodes. The incidence of ventricular arrhythmia post S1S2 stimulation was recorded. Protein levels of nonphosphorylated Cx43 and total Cx43 were evaluated by Western blot. The distribution of nonphosphorylated Cx43 was observed by confocal immunofluorescence microscopy. RESULTS: Compared with the control group, the QT interval, endocardial action potential duration, transmural repolarization dispersion (TDR) and incidence of ventricular arrhythmia were significantly increased and nonphosphorylated Cx43 expression was significantly upregulated in the LPA group. Compared with the LPA group, cotreatment with AAP10 can reduce the QT interval, endocardial action potential duration, TDR and incidence of ventricular arrhythmia (25.0% vs 62.5%, P < 0.01) and downregulate nonphosphorylated Cx43. CONCLUSIONS: LPA could promote the arrhythmia possibly by upregulating nonphosphorylated Cx43 and subsequent gap junction transmission inhibition. Gap junction enhancer AAP10 could attenuate the pro-arrhythmic effect of LPA probably by downregulating myocardial nonphosphorylated Cx43 expression.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/metabolismo , Lisofosfolipídeos/efeitos adversos , Oligopeptídeos/farmacologia , Animais , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia , Conexina 43/metabolismo , Coelhos
10.
Pain ; 152(3): 643-648, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21239112

RESUMO

Sphingosine-1-phosphate (S1P) is an important mediator of inflammation recently shown in in vitro studies to increase the excitability of small-diameter sensory neurons, at least in part, via activation of the S1P(1) receptor subtype. Activation of S1PR(1) has been reported to increase the formation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-derived superoxide (O(2)(·-)) and nitric oxide synthase (NOS)-derived nitric oxide (NO). This process favors the formation of peroxynitrite (ONOO(-) [PN]), a potent mediator of hyperalgesia associated with peripheral and central sensitization. The aims of our study were to determine whether S1P causes peripheral sensitization and thermal hyperalgesia via S1PR(1) activation and PN formation. Intraplantar injection of S1P in rats led to a time-dependent development of thermal hyperalgesia that was blocked by the S1PR(1) antagonist W146, but not its inactive enantiomer W140. The hyperalgesic effects of S1P were mimicked by intraplantar injection of the well-characterized S1PR(1) agonist SEW2871. The development of S1P-induced hyperalgesia was blocked by apocynin, a NADPH oxidase inhibitor; N(G)-nitro-l-arginine methyl ester, a nonselective NOS inhibitor; and by the potent PN decomposition catalysts (FeTM-4-PyP(5+) and MnTE-2-PyP(5+)). Our findings provide mechanistic insight into the signaling pathways engaged by S1P in the development of hyperalgesia and highlight the contribution of the S1P(1) receptor-to-PN signaling in this process. Sphingosine-1-phosphate (S1P)-induced hyperalgesia is mediated by S1P1 receptor activation and mitigated by inhibition or decomposition of peroxynitrite, providing a target pathway for novel pain management strategies.


Assuntos
Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Lisofosfolipídeos/efeitos adversos , Ácido Peroxinitroso/metabolismo , Esfingosina/análogos & derivados , Acetofenonas/uso terapêutico , Anilidas/uso terapêutico , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Masculino , Metaloporfirinas/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Organofosfonatos/uso terapêutico , Oxidiazóis/efeitos adversos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Esfingosina/efeitos adversos , Tiofenos/efeitos adversos , Fatores de Tempo
11.
Biopharm Drug Dispos ; 31(7): 396-406, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20623701

RESUMO

The pharmacokinetic (PK) and pharmacodynamic (PD) parameters of ONO-4641 in humans were estimated using preclinical data in order to provide essential information to better design future clinical studies. The characterization of PK/PD was measured in terms of decreased lymphocyte counts in blood after administration of ONO-4641, a sphingosine 1-phosphate receptor modulator. Using a two-compartment model, human PK parameters were estimated from preclinical PK data of cynomolgus monkey and in vitro human metabolism data. To estimate human PD parameters, the relationship between lymphocyte counts and plasma concentrations of ONO-4641 in cynomolgus monkeys was determined. The relationship between lymphocyte counts and plasma concentrations of ONO-4641 was described by an indirect-response model. The indirect-response model had an I(max) value of 0.828 and an IC(50) value of 1.29 ng/ml based on the cynomolgus monkey data. These parameters were used to represent human PD parameters for the simulation of lymphocyte counts. Other human PD parameters such as input and output rate constants for lymphocytes were obtained from the literature. Based on these estimated human PK and PD parameters, human lymphocyte counts after administration of ONO-4641 were simulated. In conclusion, the simulation of human lymphocyte counts based on preclinical data led to the acquisition of useful information for designing future clinical studies.


Assuntos
Azetidinas/farmacologia , Azetidinas/farmacocinética , Lisofosfolipídeos/farmacologia , Lisofosfolipídeos/farmacocinética , Naftalenos/farmacologia , Naftalenos/farmacocinética , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Azetidinas/efeitos adversos , Azetidinas/sangue , Simulação por Computador , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Contagem de Linfócitos , Linfócitos/efeitos dos fármacos , Lisofosfolipídeos/efeitos adversos , Lisofosfolipídeos/sangue , Macaca fascicularis , Microssomos Hepáticos/metabolismo , Naftalenos/efeitos adversos , Naftalenos/sangue , Ligação Proteica , Esfingosina/efeitos adversos , Esfingosina/sangue , Esfingosina/farmacocinética , Esfingosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...