Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Physiol ; 235(12): 9399-9413, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32356317

RESUMO

p120-catenin (p120) serves as a stabilizer of the calcium-dependent cadherin-catenin complex and loss of p120 expression has been observed in several types of human cancers. The p120-dependent E-cadherin-ß-catenin complex has been shown to mediate calcium-induced keratinocyte differentiation via inducing activation of plasma membrane phospholipase C-γ1 (PLC-γ1). On the other hand, PLC-γ1 has been shown to interact with phosphatidylinositol 3-kinase enhancer in the nucleus and plays a critical role in epidermal growth factor-induced proliferation of oral squamous cell carcinoma (OSCC) cells. To determine whether p120 suppresses OSCC proliferation and tumor growth via inhibiting PLC-γ1, we examined effects of p120 knockdown or p120 and PLC-γ1 double knockdown on proliferation of cultured OSCC cells and tumor growth in xenograft OSCC in mice. The results showed that knockdown of p120 reduced levels of PLC-γ1 in the plasma membrane and increased levels of PLC-γ1 and its signaling in the nucleus in OSCC cells and OSCC cell proliferation as well as xenograft OSCC tumor growth. However, double knockdown of p120 and PLC-γ1 or knockdown of PLC-γ1 alone did not have any effect. Immunohistochemical analysis of OSCC tissue from patients showed a lower expression level of p120 and a higher expression level of PLC-γ1 compared with that of adjacent noncancerous tissue. These data indicate that p120 suppresses OSCC cell proliferation and tumor growth by inhibiting signaling mediated by nuclear PLC-γ1.


Assuntos
Cateninas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Neoplasias Bucais/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Cálcio da Dieta/farmacologia , Carcinoma de Células Escamosas/patologia , Cateninas/metabolismo , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias Bucais/patologia , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Transdução de Sinais/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia
2.
Hum Mol Genet ; 26(16): 3144-3160, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28541476

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative disease with motor, cognitive and psychiatric impairment. Dysfunctions in HD models have been related to reduced levels of striatal brain-derived neurotrophic factor (BDNF) and imbalance between its receptors TrkB and p75(NTR). Thus, molecules with activity on the BDNF/TrkB/p75 system can have therapeutic potential. 7,8-Dihydroxyflavone (7,8-DHF) was described as a TrkB agonist in several models of neuro-degenerative diseases, however, its TrkB activation profile needs further investigation due to its pleiotropic properties and divergence from BDNF effect. To investigate this, we used in vitro and in vivo models of HD to dissect TrkB activation upon 7,8-DHF treatment. 7,8-DHF treatment in primary cultures showed phosphorylation of TrkBY816 but not TrkBY515 with activation of the PLCγ1 pathway leading to morphological and functional improvements. Chronic administration of 7,8-DHF delayed motor deficits in R6/1 mice and reversed deficits on the Novel Object Recognition Test (NORT) at 17 weeks. Morphological and biochemical analyses revealed improved striatal levels of enkephalin, and prevention of striatal volume loss. We found a TrkBY816 but not TrkBY515 phosphorylation recovery in striatum concordant with in vitro results. Additionally, 7,8-DHF normalized striatal levels of induced and neuronal nitric oxide synthase (iNOS and nNOS, respectively) and ameliorated the imbalance of p75/TrkB. Our results provide new insights into the mechanism of action of 7,8-DHF suggesting that its effect through the TrkB receptor in striatum is via selective phosphorylation of its Y816 residue and activation of PLCγ1 pathway, but pleiotropic effects of the drug also contribute to its therapeutic potential.


Assuntos
Flavonas/metabolismo , Flavonas/uso terapêutico , Doença de Huntington/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Flavonas/farmacologia , Hipocampo/metabolismo , Doença de Huntington/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Neurônios Motores/efeitos dos fármacos , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Fosforilação , Receptor trkB/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Am J Physiol Heart Circ Physiol ; 311(5): H1214-H1224, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27638877

RESUMO

We investigated the acute effects of glucagon-like peptide-1 (GLP-1), GLP-1(1-36), and GLP-1(7-36) on vascular endothelial growth factor-A (VEGFA)-induced endothelium-dependent signaling and vasodilation. Our hypothesis was that GLP-1 released from intestinal l-cells modulates processes related to PLCγ activation, Src, and endothelial NOS (eNOS) signaling, thereby controlling endothelial vessel tone. By using RT-PCR analysis, we found mRNA for the GLP-1 receptor (GLP-1R) in human dermal microvascular endothelial cells (HDMEC), human retinal microvascular endothelial cells, and rat arteries. In isolated rat mesenteric resistance arteries precontracted with the thromboxane analog U46619 to 80-90% of maximum contraction, VEGFA (25 ng/ml) caused a small and gradual relaxation (28.9 ± 3.9%). Pretreatment of arteries with either GLP-1(1-36) (500 nM) or GLP-1(7-36) (1 nM) abolished the VEGFA-induced relaxation. VEGFA-induced relaxations were also inhibited in endothelial-denuded arteries and in arteries pretreated with the nitric oxide synthase (NOS) inhibitor, Nω-nitro-l-arginine methyl ester (100 µM). In vivo studies on male Wistar rats also revealed that GLP-1(7-36) inhibited VEGFA-induced vasodilation of the same arteries. In isolated endothelial cells, GLP-1(1-36) and GLP-1(7-36) caused a reduction in VEGFA-induced phosphorylation of PLCγ. Ca2+ imaging of endothelial cells and rat mesenteric resistance arteries using fura-2, revealed that both GLP-1 analogs caused a reduction in VEGFA-induced Ca2+ signaling. GLP-1(1-36) also reduced VEGFA-induced eNOS phosphorylation in HDMEC. In conclusion, GLP-1 reduced relaxation induced by VEGFA in resistance arteries by inhibiting VEGFR2-mediated Ca2+ signaling and endothelial NO synthesis. GLP-1, on its own, also induced phosphorylation of Src and ERK1/2 that can lead to proliferation and is implicated in vessel permeability.


Assuntos
Células Endoteliais/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Artérias Mesentéricas/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Cálcio/metabolismo , Derme/irrigação sanguínea , Células Endoteliais/metabolismo , Inibidores Enzimáticos/farmacologia , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Humanos , Masculino , Artérias Mesentéricas/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Vasos Retinianos/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Cardiovasc Toxicol ; 16(1): 46-53, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25701259

RESUMO

Sunitinib is used extensively in the treatment of metastatic renal cell carcinoma and imatinib-resistant gastrointestinal stromal tumors. However, the undesirable cardiotoxic effects of sunitinib, such as congestive heart failure and hypertension, limit its use in the clinical setting. As multiple receptor tyrosine kinases are inhibited by sunitinib, it raises a question as to which target mediates sunitinib-induced cardiotoxicity. Here, we reported that the injection of fibroblast growth factor 2 (FGF2) mRNA into one- to two-cell stage embryos protected against sunitinib-induced cardiotoxicity in zebrafish. In addition, FGF2 significantly prevented sunitinib-induced cardiotoxicity in cardiomyoblast H9c2 cells, possibly via activating the PLC-γ/c-Raf/CREB pathway. Importantly, FGF2 did not compromise the antitumor activity of sunitinib in Caki-1 and OS-RC-2 renal cell carcinoma cells. Molecular docking simulations further revealed an interaction between the tyrosine kinase domain of FGF receptor 1 (FGFR1) and sunitinib. Taken together, our results clearly demonstrated that FGF2 inhibition plays an important role in sunitinib-induced cardiotoxicity both in vitro and in vivo. This study also provided a basis for further research on sunitinib-induced cardiotoxicity and may allow rational design of new sunitinib derivatives with fewer or weak cardiotoxic effects.


Assuntos
Inibidores da Angiogênese/toxicidade , Cardiotoxicidade/prevenção & controle , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Cardiopatias/induzido quimicamente , Cardiopatias/prevenção & controle , Indóis/antagonistas & inibidores , Indóis/toxicidade , Mioblastos/efeitos dos fármacos , Pirróis/antagonistas & inibidores , Pirróis/toxicidade , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Humanos , Simulação de Acoplamento Molecular , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Sunitinibe , Peixe-Zebra , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo
5.
J Cardiovasc Pharmacol ; 51(1): 45-54, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18209568

RESUMO

We have previously reported that green tea catechins displayed a potent antithrombotic effect by inhibition of platelet aggregation. In the present study, the antiplatelet and antithrombotic activities of epigallocatechin gallate (EGCG), the major catechin derived from green tea, were extensively investigated. EGCG inhibited arterial thrombus formation and U46619-, collagen-, and arachidonic acid (AA)-induced washed rabbit platelet aggregation in a concentration-dependent manner, with IC50 values of 61 +/- 3, 85 +/- 4, and 99 +/- 4 microM, respectively. In line with the inhibition of collagen-induced platelet aggregation, EGCG revealed blocking of the collagen-mediated phospholipase (PL) Cgamma2 and protein tyrosine phosphorylation, and it caused concentration-dependent decreases of cytosolic calcium mobilization, AA liberation, and serotonin secretion. In addition, the platelet aggregation, intracellular Ca2+ mobilization, and protein tyrosine phosphorylation induced by thapsigargin, a Ca2(+)-ATPase pump inhibitor, were completely blocked by EGCG. Contrary to the inhibition of AA-induced platelet aggregation, EGCG failed to inhibit cyclooxygenase and thromboxane (TX) A2 synthase activities, but it concentration-dependently elevated AA-mediated PGD2 formation. In contrast, epigallocatechin (EGC), a structural analogue of EGCG lacking a galloyl group in the 3' position, slightly inhibited collagen-stimulated cytosolic calcium mobilization, but failed to affect other signal transductions as did EGCG in activated platelets and arterial thrombus formation. These results suggest that antiplatelet activity of EGCG may be attributable to its modulation of multiple cellular targets, such as inhibitions of PLCgamma2, protein tyrosine phosphorylation and AA liberation, and elevation of cellular PGD2 levels, as well as maintaining Ca2(+)-ATPase activity, which may underlie its beneficial effect on the atherothrombotic diseases.


Assuntos
Catequina/análogos & derivados , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Chá/química , Animais , Ácido Araquidônico/metabolismo , ATPases Transportadoras de Cálcio/efeitos dos fármacos , ATPases Transportadoras de Cálcio/metabolismo , Catequina/administração & dosagem , Catequina/farmacologia , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Masculino , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Fosforilação , Inibidores da Agregação Plaquetária/administração & dosagem , Prostaglandina D2/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley
6.
Exp Cell Res ; 314(3): 530-42, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18054784

RESUMO

Autotaxin (ATX) is a potent tumor cell motogen that can produce lysophosphatidic acid (LPA) from lysophosphatidylcholine. LPA is a lipid mediator that has also been shown to modulate tumor cell invasion. Autotaxin mRNA is expressed at significant levels in the intestine. Likewise, LPA2 receptor levels have been shown to be elevated in colon cancers. The molecular mechanism of ATX/LPA-induced increase in intestinal cell migration however, remains poorly understood. Villin is an intestinal and renal epithelial cell specific actin regulatory protein that modifies epithelial cell migration. In this study we demonstrate that both Caco-2 (endogenous villin) and MDCK (exogenous villin) cells, which express primarily LPA2 receptors, show enhanced cell migration in response to ATX/LPA. ATX and LPA treatment results in the rapid formation of lamellipodia and redistribution of villin to these cell surface structures, suggesting a role for villin in regulating this initial event of cell locomotion. The LPA-induced increase in cell migration required activation of c-src kinase and downstream tyrosine phosphorylation of villin by c-src kinase. LPA stimulated cell motility was determined to be insensitive to pertussis toxin, but was regulated by activation of PLC-gamma 1. Together, our results show that in epithelial cells ATX and LPA act as strong stimulators of cell migration by recruiting PLC-gamma 1 and villin, both of which participate in the initiation of protrusion.


Assuntos
Movimento Celular/fisiologia , Mucosa Intestinal/metabolismo , Lisofosfolipídeos/fisiologia , Proteínas dos Microfilamentos/metabolismo , Complexos Multienzimáticos/fisiologia , Fosfodiesterase I/fisiologia , Pseudópodes/metabolismo , Pirofosfatases/fisiologia , Actinas/metabolismo , Animais , Células CACO-2 , Carcinoma/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Cães , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Neoplasias Intestinais/metabolismo , Lisofosfolipídeos/farmacologia , Proteínas dos Microfilamentos/efeitos dos fármacos , Proteínas dos Microfilamentos/farmacologia , Complexos Multienzimáticos/farmacologia , Fosfodiesterase I/farmacologia , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Diester Fosfórico Hidrolases , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Pseudópodes/efeitos dos fármacos , Pseudópodes/ultraestrutura , Pirofosfatases/farmacologia , Receptores de Ácidos Lisofosfatídicos/efeitos dos fármacos , Receptores de Ácidos Lisofosfatídicos/metabolismo , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
7.
Brain Res ; 1156: 9-20, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17524370

RESUMO

Alterations in neurotrophic signaling pathways may contribute to the changes in the mesolimbic dopamine system induced by chronic morphine exposure. In a rat model of morphine dependence, we previously identified increased levels of phospholipase C gamma-1 (PLCgamma1) immunoreactivity specifically within the ventral tegmental area (VTA) following chronic morphine treatment. Using an antibody specific for the tyrosine-phosphorylated, activated form of PLCgamma1, we now show that chronic morphine also significantly upregulates PLCgamma1 activity in the VTA, as well as in the nucleus accumbens and hippocampus, regions which are also implicated in the reinforcing properties of morphine. In contrast, no increase in PLCgamma1 activity was found in the substantia nigra or dorsal striatum. HSV-mediated overexpression of PLCgamma1 in PC12 cells induced ERK activation via a mechanism dependent, in part, on both MAP-ERK kinase (MEK) and protein kinase C. PLCgamma1 overexpression in the VTA similarly induced ERK activation in the VTA in vivo. As chronic morphine treatment has been shown to increase ERK activity within the VTA, the current results suggest that increased PLCgamma1 activity may be an upstream mediator of this effect.


Assuntos
Corpo Estriado/enzimologia , Morfina/farmacologia , Neurônios/enzimologia , Fosfolipase C gama/metabolismo , Animais , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Vetores Genéticos , Cinética , Sistema Límbico/enzimologia , Fatores de Crescimento Neural/metabolismo , Células PC12 , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/genética , Fosforilação , Ratos , Proteínas Recombinantes/metabolismo , Simplexvirus
8.
J Cardiovasc Pharmacol ; 49(5): 280-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17513946

RESUMO

Hyperproliferation of platelet-derived growth factor (PDGF)-BB-induced vascular smooth muscle cell (VSMC) is a hallmark of atherosclerosis and related vascular disorders. In the previous study, we reported that KTJ740 [2-chloro-3-(4-(ethylcarboxy)-phenyl)-amino-1,4-naphthoquinone], a newly synthesized vitamin K derivative, has potent antithrombotic effects in mice and antiplatelet activity in vitro and ex vivo. In the present study, we have tested that KTJ740 could inhibit PDGF-BB-stimulated VSMC proliferation. We have examined the potential inhibitory effect of this compound on rat aortic smooth muscle cells (RASMCs). Our results show that this compound significantly inhibits PDGF-BB-stimulated RASMC number and DNA synthesis in a concentration-dependent manner. Furthermore, we have examined its effect on cell cycle progression by flow cytometry. KTJ740 treatment resulted in a significant arrest in cell cycle progression of RASMCs induced by PDGF-BB, and this effect was achieved by suppressing activation of PDGF-beta receptor (PDGF-Rbeta) tyrosine kinase pathway. These results suggest that a possibility of KTJ740 can be a potential agent to control vascular disorders and its antiproliferative mechanism may be mediated through PDGF-Rbeta tyrosine kinase-dependent signaling pathway.


Assuntos
Indutores da Angiogênese/farmacologia , Aorta/citologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fibrinolíticos/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Naftoquinonas/farmacologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Análise de Variância , Animais , Becaplermina , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Fosfolipase C gama/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Receptores do Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
9.
Exp Hematol ; 35(3): 368-73, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17309817

RESUMO

OBJECTIVES: Phospholipase C (PLC) gamma 1 has been shown to mediate signal transduction of tyrosine kinases and affect function of hematopoietic cells. However, its role in hematopoiesis during embryonic development is currently unclear. In this study, we examined this issue using morpholino (MO) gene knockdown in zebrafish embryos METHODS: MO targeting at the exon-1-intron-1 junction of zebrafish PLC-gamma1 was injected into embryos at the one- to four-cell stage (referred herein zPLC-gamma1(MO) embryos). Primitive hematopoiesis was examined quantitatively by flow cytometry in Tg(gata1:GFP) embryos and by real-time quantitative polymerase chain reaction at 18 hours-post-fertilization (hpf), before the onset of circulation. The embryos were also treated with receptor inhibitors of vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor at 25, 1, and 30 micromol/L, respectively, from one cell until 48 hpf. RESULTS: Erythropoiesis was reduced in zPLC-gamma1(MO) embryos, as shown by the reduction in gata1(+) cells (wild-type: 4.32% +/- 0.10% vs zPLC-gamma1(MO): 2.38% +/- 0.11%, p = 0.021) and gata1 and alpha-embryonic hemoglobin expression [0.47 +/- 0.06-fold (p = 0.013) and 0.46 +/- 0.04-fold (p = 0.013)]. Expression of scl, lmo-2 (early hematopoietic progenitors), pu.1, and l-plastin (myelomonocytic lineages) as well as fli1 (vascular progenitors) were unaffected. Fli1(+) cells in Tg(fli1:GFP) embryos were also unaffected by zPLC-gamma1(MO). When embryos were incubated with receptor inhibitors of vascular endothelial growth factor (VEGFRTKI), fibroblast growth factor (SU5402), or platelet-derived growth factor (AG1296), only VEGFRTKI reduced erythropoiesis [VEGFRTKI: 2.10% +/- 0.07% (p = 0.021) vs SU5402: 4.08% +/- 0.12% (p = 0.248) vs AG1296: 4.12% +/- 0.14% (p = 0.149)]. CONCLUSION: PLC-gamma1 is involved in the regulation of primitive hematopoiesis in zebrafish embryos, which is distinct from its later effect on vascular formation.


Assuntos
Hematopoese/fisiologia , Fosfolipase C gama/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia , Animais , Eritropoese/efeitos dos fármacos , Eritropoese/fisiologia , Fator 1 de Crescimento de Fibroblastos/administração & dosagem , Citometria de Fluxo/métodos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes/métodos , Proteínas de Fluorescência Verde/genética , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Oligonucleotídeos Antissenso/administração & dosagem , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/genética , Fator de Crescimento Derivado de Plaquetas/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Peixe-Zebra/genética
10.
J Leukoc Biol ; 81(5): 1276-86, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17284681

RESUMO

Chinese herbs are useful edible and medicinal plants for their immune modulatory functions. We have proven that (S)-armepavine (C19H23O3N; MW313) from Nelumbo nucifera inhibits the proliferation of human PBMCs activated with PHA and improves autoimmune diseases in MRL/MpJ-lpr/lpr mice. In the present study, the pharmacological activities of (S)-armepavine were evaluated in PHA-activated PBMCs. The results showed that (S)-armepavine suppressed PHA-induced PBMC proliferation and genes expression of IL-2 and IFN-gamma without direct cytotoxicity. Inhibition of NF-AT and NF-kappaB activation suggested phospholipase Cgamma (PLCgamma)-mediated Ca2+ mobilization and protein kinase C activation were blocked by (S)-armepavine. Phosphorylation of PLCgamma is regulated by lymphocyte-specific kinase (Lck), ZAP-70, and IL-2-inducible T cell kinase (Itk). We found (S)-armepavine inhibited PHA-induced phosphorylation of Itk and PLCgamma efficiently but did not influence Lck or ZAP-70 phosphorylation. In addition, ZAP-70-mediated pathways, such as the association of linker for activation of T cells with PLCgamma and activation of ERK, were also intact in the presence of (S)-armepavine. Finally, reduction of phosphoinositide 3,4,5-trisphosphate formation and Akt phosphorylation suggested that (S)-armepavine inhibited Itk, and PLCgamma phosphorylation might be a result of the influence of PI-3K activation. Addition of exogenous IL-2 or PMA/A23187 rescued PBMC proliferation in the presence of (S)-armepavine. Therefore, we concluded that (S)-armepavine inhibited PHA-induced cell proliferation and cytokine production in a major way by blocking membrane-proximal effectors such as Itk and PLCgamma in a PI-3K-dependent manner.


Assuntos
Alcaloides/farmacologia , Benzilisoquinolinas/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama/efeitos dos fármacos , Proteínas Tirosina Quinases/efeitos dos fármacos , Alcaloides/química , Alcaloides/isolamento & purificação , Benzilisoquinolinas/química , Benzilisoquinolinas/isolamento & purificação , Calcimicina/farmacologia , Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Interleucina-2/antagonistas & inibidores , Interleucina-2/biossíntese , Leucócitos Mononucleares/imunologia , Conformação Molecular , Peso Molecular , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Nelumbo/química , Fosfatidilinositol 3-Quinases/biossíntese , Inibidores de Fosfoinositídeo-3 Quinase , Fosfolipase C gama/metabolismo , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Sementes/química , Relação Estrutura-Atividade , Acetato de Tetradecanoilforbol/farmacologia
11.
Neuropsychopharmacology ; 32(10): 2152-62, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17314919

RESUMO

Previous studies suggest that brain-derived neurotrophic factor and its receptor TrkB are critically involved in the therapeutic actions of antidepressant drugs. We have previously shown that the antidepressants imipramine and fluoxetine produce a rapid autophosphorylation of TrkB in the rodent brain. In the present study, we have further examined the biochemical and functional characteristics of antidepressant-induced TrkB activation in vivo. We show that all the antidepressants examined, including inhibitors of monoamine transporters and metabolism, activate TrkB rapidly in the rodent anterior cingulate cortex and hippocampus. Furthermore, the results indicate that acute and long-term antidepressant treatments induce TrkB-mediated activation of phospholipase-Cgamma1 (PLCgamma1) and increase the phosphorylation of cAMP-related element binding protein, a major transcription factor mediating neuronal plasticity. In contrast, we have not observed any modulation of the phosphorylation of TrkB Shc binding site, phosphorylation of mitogen-activated protein kinase or AKT by antidepressants. We also show that in the forced swim test, the behavioral effects of specific serotonergic antidepressant citalopram, but not those of the specific noradrenergic antidepressant reboxetine, are crucially dependent on TrkB signaling. Finally, brain monoamines seem to be critical mediators of antidepressant-induced TrkB activation, as antidepressants reboxetine and citalopram do not produce TrkB activation in the brains of serotonin- or norepinephrine-depleted mice. In conclusion, our data suggest that rapid activation of the TrkB neurotrophin receptor and PLCgamma1 signaling is a common mechanism for all antidepressant drugs.


Assuntos
Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Fosfolipase C gama/efeitos dos fármacos , Receptor trkB/agonistas , Transdução de Sinais/efeitos dos fármacos , Inibidores da Captação Adrenérgica/farmacologia , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Encéfalo/metabolismo , Química Encefálica/efeitos dos fármacos , Química Encefálica/fisiologia , Citalopram/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/metabolismo , Transtorno Depressivo/fisiopatologia , Giro do Cíngulo/efeitos dos fármacos , Giro do Cíngulo/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfolinas/farmacologia , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Reboxetina , Receptor trkB/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
12.
Tumour Biol ; 27(4): 201-10, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16651854

RESUMO

BACKGROUND: HER2, which is overexpressed in 25-30% of human breast cancers, is a tyrosine kinase receptor critical for the signal transduction network that regulates proliferation, migration and apoptosis of cells. METHOD: We report the effects of two novel HER2-binding affibody molecules (Affibody), (ZHER2:4)2 and ZHER2:342, on intracellular signal transduction pathways (Erk1/2, Akt and PLCgamma1) using quantitative immunoblotting techniques and their biological effects in cell culture. The clinically approved antibody trastuzumab (Herceptin) was used as reference substance. RESULTS: Our data showed that, although all substances target HER2, the effects on the receptor and signaling molecules differed. For example, HER2 phosphorylation was induced by trastuzumab and (ZHER2:4)2 but inhibited by ZHER2:342. The effects these substances had on signal transduction correlated to some degree with changes in growth and migration, e.g. (ZHER2:4)2 stimulated phosphorylation of Erk1/2 and PLCgamma1, as well as growth and migration, while ZHER2:342 did not. ZHER2:342 even inhibited phosphorylation of PLCgamma1 and migration. CONCLUSION: Our data suggest that ZHER2:342 is a promising small agent (7 kDa) that may be used as an alternative, or complement, to trastuzumab. If radiolabelled, it can hopefully also be used for HER2 imaging and radionuclide therapy.


Assuntos
Receptor ErbB-2/metabolismo , Transdução de Sinais/fisiologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Divisão Celular , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Ovarianas , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Receptor ErbB-2/genética , Transdução de Sinais/efeitos dos fármacos , Trastuzumab
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...