Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Pharmacol Res ; 203: 107173, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38580186

RESUMO

Our recent multi-omics studies have revealed rich sources of novel bioactive proteins and polypeptides from marine organisms including cnidarians. In the present study, we initially conducted a transcriptomic analysis to review the composition profile of polypeptides from Zoanthus sociatus. Then, a newly discovered NPY-like polypeptide-ZoaNPY was selected for further in silico structural, binding and virtually pharmacological studies. To evaluate the pro-angiogenic effects of ZoaNPY, we employed an in vitro HUVECs model and an in vivo zebrafish model. Our results indicate that ZoaNPY, at 1-100 pmol, enhances cell survival, migration and tube formation in the endothelial cells. Besides, treatment with ZoaNPY could restore a chemically-induced vascular insufficiency in zebrafish embryos. Western blot results demonstrated the application of ZoaNPY could increase the phosphorylation of proteins related to angiogenesis signaling including PKC, PLC, FAK, Src, Akt, mTOR, MEK, and ERK1/2. Furthermore, through molecular docking and surface plasmon resonance (SPR) verification, ZoaNPY was shown to directly and physically interact with NPY Y2 receptor. In view of this, all evidence showed that the pro-angiogenic effects of ZoaNPY involve the activation of NPY Y2 receptor, thereby activating the Akt/mTOR, PLC/PKC, ERK/MEK and Src- FAK-dependent signaling pathways. Furthermore, in an excision wound model, the treatment with ZoaNPY was shown to accelerate the wound healing process in mice. Our findings provide new insights into the discovery and development of novel pro-angiogenic drugs derived from NPY-like polypeptides in the future.


Assuntos
Cnidários , Peptídeos , Receptores de Neuropeptídeo Y , Animais , Humanos , Camundongos , Movimento Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Ligantes , Simulação de Acoplamento Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Neuropeptídeo Y/farmacologia , Peptídeos/farmacologia , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Receptores de Neuropeptídeo Y/efeitos dos fármacos , Receptores de Neuropeptídeo Y/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo , Peixe-Zebra , Cnidários/química , Fosfoinositídeo Fosfolipase C/efeitos dos fármacos , Fosfoinositídeo Fosfolipase C/metabolismo
2.
Pak J Pharm Sci ; 34(3): 891-898, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34602411

RESUMO

This study investigates the effects of ß-carboline alkaloids from Peganum harmala on FAK/PI3K/AKT/mTOR pathway in gastric cancer cell line SGC-7901 and tumor-bearing mice. Western blot, immunohistochemistry and RT-PCR were performed to detect protein and mRNA expressions of BCL-2, Bax, FAK, PI3K, AKT and mTOR. Mice model of gastric tumor was established with SGC-7901 cells. TUNEL assay was used to detect apoptosis. HE staining was used to observe morphological changes. In vitro, the protein and mRNA expressions of FAK, PI3K, AKT and mTOR in ß-carboline alkaloids groups were significantly lower than those in control and fluorouracil groups (P<0.05). BCL-2 decreased while Bax increased. In vivo, the tumor weights of ß-carboline alkaloids and fluorouracil groups were significantly lower than those of control group (P<0.05). FAK, PI3K, AKT and mTOR proteins in tumor tissues of ß-carboline alkaloids and fluorouracil groups were significantly lower than control group (P<0.05). Additionally, ß-carboline alkaloids treatment in vivo caused obvious cell necrosis and apoptosis. Conclusively, ß-carboline alkaloids can reduce FAK, PI3K, AKT and mTOR expressions at both protein and mRNA levels in SGC-7901 cells and tumor tissues formed by SGC-7901 cells. They may be targets of ß-carboline in FAK/PI3K/AKT/mTOR pathway.


Assuntos
Alcaloides/farmacologia , Carbolinas/farmacologia , Carcinoma/metabolismo , Quinase 1 de Adesão Focal/efeitos dos fármacos , Peganum , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Serina-Treonina Quinases TOR/efeitos dos fármacos , Animais , Carcinoma/genética , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Humanos , Camundongos , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Gástricas/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
3.
Invest New Drugs ; 39(2): 377-385, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32989643

RESUMO

Purpose Our study aimed to investigate the antitumor effects of rAj-Tspin on BEL-7402 hepatocellular carcinoma cells and to explore the underlying mechanism. Method For the in vivo experiment, BEL-7402 cells were inoculated subcutaneously into the axilla of nude mice to generate a BEL-7402 cell-bearing model, and model mice were then treated with different doses of rAj-Tspin. A CCK-8 assay was used to evaluate the in vitro viability of BEL-7402 and LO2 cells after treatment with different concentrations of rAj-Tspin. The effects of rAj-Tspin on BEL-7402 cell apoptosis, migration and invasion were evaluated by AO/EB and Hoechst fluorescent staining and by scratch and Transwell assays, and the tumor-suppressive mechanism of rAj-Tspin was explored by Western blotting. Results rAj-Tspin suppressed the proliferation of BEL-7402 cells with an IC50 of 0.89 µM. The results of both microscopic analysis and Western blotting suggested that rAj-Tspin induced the apoptosis of BEL-7402 cells through a mitochondria-dependent pathway. Furthermore, rAj-Tspin disrupted EMT; this disruption ultimately caused BEL-7402 cells to lose their shape and decreased their migration and invasion. Moreover, rAj-Tspin might inhibit the proliferation and metastasis of BEL-7402 cells through the ITGB1-FAK-AKT pathway. Conclusion rAj-Tspin exerts an antitumor effect through the ITGB1-FAK-Akt signaling pathway and exhibits low toxicity at an effective dose.


Assuntos
Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Stichopus , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Quinase 1 de Adesão Focal/efeitos dos fármacos , Integrina beta1/efeitos dos fármacos , Neoplasias Hepáticas/patologia , Masculino , Toxinas Marinhas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/prevenção & controle , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Cell Physiol ; 235(11): 8334-8344, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32239671

RESUMO

E-cadherin, a central component of the adherens junction (AJ), is a single-pass transmembrane protein that mediates cell-cell adhesion. The loss of E-cadherin surface expression, and therefore cell-cell adhesion, leads to increased cell migration and invasion. Treatment of colorectal cancer (CRC)-derived cells (SW-480 and HT-29) with 2.0 mM metformin promoted a redistribution of cytosolic E-cadherin to de novo formed puncta along the length of the contacting membranes of these cells. Metformin also promoted translocation from the cytosol to the plasma membrane of p120-catenin, another core component of the AJs. Furthermore, E-cadherin and p120-catenin colocalized with ß-catenin at cell-cell contacts. Western blot analysis of lysates of CRC-derived cells revealed a substantial metformin-induced increase in the level of p120-catenin as well as E-cadherin phosphorylation on Ser838/840 , a modification associated with ß-catenin/E-cadherin interaction. These modifications in E-cadherin, p120-catenin and ß-catenin localization suggest that metformin induces rebuilding of AJs in CRC-derived cells. Those modifications were accompanied by the inhibition of focal adhesion kinase (FAK), as revealed by a significant decrease in the phosphorylation of FAK at Tyr397 and paxillin at Tyr118 . These changes were associated with a reduction in the numbers, but an increase in the size, of focal adhesions and by the inhibition of cell migration. Overall, these observations indicate that metformin targets multiple pathways associated with CRC development and progression.


Assuntos
Junções Aderentes/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Quinase 1 de Adesão Focal/metabolismo , Metformina/farmacologia , Junções Aderentes/metabolismo , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Quinase 1 de Adesão Focal/efeitos dos fármacos , Humanos , Transporte Proteico/efeitos dos fármacos
5.
J Med Chem ; 62(5): 2508-2520, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30739444

RESUMO

Focal adhesion tyrosine kinase (PTK2) is often overexpressed in human hepatocellular carcinoma (HCC), and several reports have linked PTK2 depletion and/or pharmacological inhibition to reduced tumorigenicity. However, the clinical relevance of targeting PTK2 still remains to be proven. Here, we present two highly selective and functional PTK2 proteolysis-targeting chimeras utilizing von Hippel-Lindau and cereblon ligands to hijack E3 ligases for PTK2 degradation. BI-3663 (cereblon-based) degrades PTK2 with a median DC50 of 30 nM to >80% across a panel of 11 HCC cell lines. Despite effective PTK2 degradation, these compounds did not phenocopy the reported antiproliferative effects of PTK2 depletion in any of the cell lines tested. By disclosing these compounds, we hope to provide valuable tools for the study of PTK2 degradation across different biological systems.


Assuntos
Quinase 1 de Adesão Focal/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Recombinantes/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Humanos , Ligantes , Proteólise , Interferência de RNA
6.
Life Sci ; 213: 248-257, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30292831

RESUMO

PURPOSE: Statins extended their hypocholestremic effect to show a promising anticancer activity. Hepatocellular carcinoma (HCC), the third common cause of cancer-related death, responded positively to statins. Some in-vitro studies reveal the rosuvastatin antitumor effect, but barely in-vivo studies. Hence, we evaluated the antitumor potential of rosuvastatin in a HCC model, the possible signaling cues involved, and whether it augments the dasatinib anticancer effect. METHOD: For the in-vitro study, the IC50 and the combination (CI)/dose reduction (DRI) indices were determined for HCC cell line (HepG2) treated with dasatinib and/or rosuvastatin. For the in-vivo study, mice with diethylnitrosamine-induced HCC were treated for 21 days with dasatinib and/or rosuvastatin (10 and 20 mg/kg, respectively). The p-focal adhesion kinase/p-rous sarcoma oncogene cellular homolog (p-FAK/p-Src) cascade and its downstream molecules were assessed. RESULTS: The in-vitro study confirmed the synergistic effect of rosuvastatin with dasatinib, which entailed the in-vivo results. The two drugs decreased the p-FAK/p-Src cue along with p-Ras/c-Raf, p-STAT-3, and p-Akt levels to enhance apoptosis by an increase in caspase-3 level and a decline in survivin level. Additionally, they inhibited HGF, VEGF, and the MMP-9. Moreover, the different treatments downregulated the expression of proliferative cell nuclear antigen (PCNA) and Ki-67. The best effect was mediated by the combination regimen that surpassed the effect of either drug alone. CONCLUSION: Our results highlighted some of the signals involved in rosuvastatin antitumor effect and nominate it as an adds-on therapy with dasatinib to yield a better effect in HCC through inhibiting the FAK/Src cascade.


Assuntos
Dasatinibe/farmacologia , Rosuvastatina Cálcica/farmacologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Quinase 1 de Adesão Focal/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/efeitos dos fármacos
7.
Oncotarget ; 8(13): 21674-21691, 2017 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28423510

RESUMO

Genistein is one of the main components of soy-based foods, which are widely known for their many benefits, including anti-cancer, anti-inflammatory, and antioxidant effects. In this study, we investigated the anti-metastasis effects of genistein on B16F10 melanoma cells. Our results showed that genistein strongly inhibited B16F10 cell proliferation and induced apoptosis in time- and concentration-dependent manners. Genistein altered the morphology of B16F10 cells to an elongated shape with slim pseudopodia-like protrusions. Moreover, genistein inhibited the invasion and migration abilities of B16F10 cells in a dose-dependent manner. On one hand, a high concentration of genistein (100 µM) significantly inhibited cell adhesion and migration, as shown by wound healing assays and transwell-migration and invasion assays. Furthermore, the expression levels of p-FAK, p-paxillin, tensin-2, vinculin, and α-actinin were decreased by genistein. As a result, genistein is believed to strongly downregulate the migration and invasion abilities of B16F10 cells via the FAK/paxillin pathway. Moreover, p-p38, p-ERK, and p-JNK levels were also dramatically decreased by treatment with genistein. Finally, genistein significantly decreased the gene expression of FAK, paxillin, vimentin, and epithelial-to-mesenchymal transition-related transcription factor Snail, as shown by real-time PCR (qPCR) analysis. On the other hand, a lower concentration of genistein (12.5 µM) significantly promoted both invasion and migration by activating the FAK/paxillin and MAPK signaling cascades. Taken together, this study showed for the first time that genistein exerts dual functional effects on melanoma cells. Our findings suggest that genistein regulates the FAK/paxillin and MAPK signaling pathways in a highly concentration-dependent manner. Patients with melanoma should therefore be cautious of consuming soy-based foods in their diets.


Assuntos
Anticarcinógenos/farmacologia , Genisteína/farmacologia , Melanoma Experimental/patologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Imunofluorescência , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Marcação In Situ das Extremidades Cortadas , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Invasividade Neoplásica/patologia , Paxilina/efeitos dos fármacos , Paxilina/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
8.
World J Urol ; 35(8): 1247-1254, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28013345

RESUMO

OBJECTIVES: To investigate the effect of simulated physiological stretch on the expression of extracellular matrix (ECM) proteins and the role of integrin α4/αv, focal adhesion kinase (FAK), extracellular regulated protein kinases 1/2 (ERK1/2) in the stretch-induced ECM protein expression of human bladder smooth muscle cells (HBSMCs). METHODS: HBSMCs were seeded onto silicone membrane and subjected to simulated physiological stretch at the range of 5, 10, and 15% elongation. Expression of primary ECM proteins in HBSMCs was analyzed by real-time polymerase chain reaction and Western blot. Specificity of the FAK and ERK1/2 was determined by Western blot with FAK inhibitor and ERK1/2 inhibitor (PD98059). Specificity of integrin α4 and integrin αv was determined with small interfering ribonucleic acid (siRNA) transfection. RESULTS: The expression of collagen I (Col1), collagen III (Col3), and fibronectin (Fn) was increased significantly under the simulated physiological stretch of 10 and 15%. Integrin α4 and αv, FAK, ERK1/2 were activated by 10% simulated physiological stretch compared with the static condition. Pretreatment of ERK1/2 inhibitor, FAK inhibitor, integrin α4 siRNA, or integrin αv siRNA reduced the stretch-induced expression of ECM proteins. And FAK inhibitor decreased the stretch-induced ERK1/2 activity and ECM protein expression. Integrin α4 siRNA or integrin αv siRNA inhibited the stretch-induced activity of FAK. CONCLUSION: Simulated physiological stretch increases the expression of ECM proteins in HBSMCs, and integrin α4/αv-FAK-ERK1/2 signaling pathway partly modulates the mechano-transducing process.


Assuntos
Proteínas da Matriz Extracelular/genética , Quinase 1 de Adesão Focal/genética , Integrina alfa4/genética , Integrina alfaV/genética , Sistema de Sinalização das MAP Quinases/genética , Miócitos de Músculo Liso/metabolismo , Fenômenos Biomecânicos , Western Blotting , Colágeno Tipo I/efeitos dos fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo III/efeitos dos fármacos , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Proteínas da Matriz Extracelular/efeitos dos fármacos , Proteínas da Matriz Extracelular/metabolismo , Fibronectinas/efeitos dos fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Flavonoides/farmacologia , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Humanos , Integrina alfa4/efeitos dos fármacos , Integrina alfa4/metabolismo , Integrina alfaV/efeitos dos fármacos , Integrina alfaV/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Bexiga Urinária/citologia
9.
Eur J Oral Sci ; 123(4): 249-53, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26058789

RESUMO

Since focal adhesion kinase (FAK) was proposed as a mediator of the inflammatory response, we have investigated the role of this molecule in the release of inflammatory cytokines by cultured human periodontal ligament fibroblasts (HPDLFs), cells that are thought to be important in the patient's response to periodontal infection. Human periodontal ligament fibroblasts were stimulated by tumor necrosis factor alpha (TNF-α) and its effects on interleukin (IL)-6 and IL-8 release were measured by ELISA. Expression of matrix metalloproteinase 2 (MMP-2) protein was analysed by western blotting. The levels of IL6, IL8, and MMP2 mRNA were evaluated by real-time PCR. Tumor necrosis factor alpha dose-dependently induced the phosphorylation of FAK, whereas small interfering FAK (siFAK) inhibited TNF-α-induced FAK phosphorylation. Tumor necrosis factor alpha also stimulated the production of IL-6, IL-8, and MMP-2 in a dose-dependent manner. Knockdown of FAK significantly suppressed TNF-α-induced expression of IL6 and IL8 mRNA and release of IL-6 and IL-8 protein in HPDLFs. Similarly, MMP-2 down-regulation was significantly prevented by siFAK. Our results strongly suggest that knockdown of FAK can decrease the production of TNF-α-induced IL-6, IL-8, and MMP-2 in HPDLFs. These effects may help in understanding the mechanisms that control expression of inflammatory cytokines in the pathogenesis of periodontitis.


Assuntos
Fibroblastos/efeitos dos fármacos , Quinase 1 de Adesão Focal/efeitos dos fármacos , Interleucina-6/análise , Interleucina-8/efeitos dos fármacos , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Ligamento Periodontal/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/enzimologia , Quinase 1 de Adesão Focal/genética , Técnicas de Silenciamento de Genes , Humanos , Ligamento Periodontal/citologia , Ligamento Periodontal/enzimologia , Fosforilação , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/administração & dosagem
10.
J Dent Res ; 94(6): 853-62, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25724555

RESUMO

Mineral trioxide aggregate is the currently recommended material of choice for clinical pulp repair despite several disadvantages, including handling inconvenience. Little is known about the signaling mechanisms involved in bioceramic-mediated dental pulp repair-particularly, dental pulp cell (DPC) migration. This study evaluated the effects of iRoot BP Plus, a novel ready-to-use nanoparticulate bioceramic putty, on DPC migration in vitro and pulp repair in vivo, focusing on possible involvement of fibroblast growth factor receptor (FGFR)-related signaling, including mitogen-activated protein kinase and Akt pathways. Treatment with iRoot BP Plus extracts enhanced horizontal and vertical migration of DPCs, which was comparable with the effects induced by mineral trioxide aggregate extracts. The DPCs exposed to iRoot BP Plus extracts demonstrated no evident apoptosis. Importantly, treatment with iRoot BP Plus extracts resulted in rapid activation of FGFR, p38 mitogen-activated protein kinase, extracellular signal-regulated kinase (ERK) 1/2, c-Jun-N-terminal kinase (JNK), and Akt signaling in DPCs. Confocal immunofluorescence staining revealed that iRoot BP Plus stimulated focal adhesion formation and stress fiber assembly in DPCs, in addition to upregulating the expression of focal adhesion molecules, including p-focal adhesion kinase, p-paxillin, and vinculin. Moreover, activation of FGFR, ERK, JNK, and Akt were found to mediate the upregulated expression of focal adhesion molecules, stress fiber assembly, and enhanced DPC migration induced by iRoot BP Plus. Consistent with the in vitro results, we observed induction of homogeneous dentin bridge formation and expression of p-focal adhesion kinase, p-FGFR, p-ERK 1/2, p-JNK, and p-Akt near injury sites by iRoot BP Plus in an in vivo pulp repair model. These data demonstrate that iRoot BP Plus can promote DPC migration and pulp repair involving the FGFR-mediated ERK 1/2, JNK, and Akt pathways. These findings provide valuable insights into the signaling mechanisms underlying nanoparticulate bioceramic-mediated pulp repair.


Assuntos
Compostos de Cálcio/farmacologia , Cerâmica/química , Polpa Dentária/citologia , Agentes de Capeamento da Polpa Dentária e Pulpectomia/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Silicatos/farmacologia , Compostos de Alumínio/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Polpa Dentária/efeitos dos fármacos , Dentina Secundária/efeitos dos fármacos , Dentinogênese/efeitos dos fármacos , Combinação de Medicamentos , Quinase 1 de Adesão Focal/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Teste de Materiais , Proteínas de Membrana/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Nanopartículas/química , Óxidos/farmacologia , Paxilina/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Ratos , Fibras de Estresse/efeitos dos fármacos , Vinculina/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos
11.
Cancer Prev Res (Phila) ; 8(5): 419-30, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25712051

RESUMO

The membrane-associated protein, focal adhesion kinase (FAK), modulates cell-extracellular matrix interactions and also conveys prosurvival and proliferative signals. Notably, increased intraepithelial FAK levels accompany transformation of premalignant oral intraepithelial neoplasia (OIN) to oral squamous cell carcinoma (OSCC). OIN chemoprevention is a patient-centric, optimal strategy to prevent OSCC's comorbidities and mortality. The cancer chemopreventive and synthetic vitamin A derivative, fenretinide, has demonstrated protein-binding capacities, for example, mTOR- and retinol-binding protein interactions. These studies used a continuum of human oral keratinocytes (normal-HPV E6/E7-transduced-OSCC) to assess potential fenretinide-FAK drug protein interactions and functional consequences on cellular growth regulation and motility. Molecular modeling studies demonstrated that fenretinide has approximately 200-fold greater binding affinity relative to the natural ligand (ATP) at FAK's kinase domain. Fenretinide also shows intermediate binding at FAK's FERM domain and interacts at the ATP-binding site of the closest FAK analogue, PYK2. Fenretinide significantly suppressed proliferation via induction of apoptosis and G2-M cell-cycle blockade. Fenretinide-treated cells also demonstrated F-actin disruption, significant inhibition of both directed migration and invasion of a synthetic basement membrane, and decreased phosphorylation of growth-promoting kinases. A commercially available FAK inhibitor did not suppress cell invasion. Notably, although FAK's FERM domain directs cell invasion, FAK inhibitors target the kinase domain. In addition, FAK-specific siRNA-treated cells showed an intermediate cell migration capacity; data which suggest cocontribution of the established migrating-enhancing PYK2. Our data imply that fenretinide is uniquely capable of disrupting FAK's and PYK2's prosurvival and mobility-enhancing effects and further extend fenretinide's chemopreventive contributions beyond induction of apoptosis and differentiation.


Assuntos
Carcinoma de Células Escamosas/patologia , Matriz Extracelular/efeitos dos fármacos , Fenretinida/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia , Neoplasias Bucais/patologia , Lesões Pré-Cancerosas/patologia , Anticarcinógenos/farmacologia , Movimento Celular/efeitos dos fármacos , Quimioprevenção , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Humanos , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/patologia , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
12.
J Endod ; 40(10): 1642-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25260738

RESUMO

INTRODUCTION: The objective of this study was to compare the cytotoxicity, inflammatory response, osteogenic effect, and the signaling mechanism of these biologic activities of 4 calcium compound-based root canal sealers (ie, Sealapex [Sybron Kerr, WA], apatite root sealer [ARS; Dentsply Sankin, Tokyo, Japan], MTA Fillapex [Angelus Indústria de Produtos Odontológicos S/A, Londrina, PR, Brazil], and iRoot SP [Innovative BioCreamix Inc, Vancouver, Canada]) in human periodontal ligament cells. METHODS: Cytotoxicity was assessed using the 3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide assay. Levels of inflammatory mediators were measured by enzyme-linked immunosorbent assay, reverse-transcription polymerase chain reaction, and Western blot analysis. Osteogenic potential was evaluated by alkaline phosphatase activity, alizarin red staining, and marker genes by reverse-transcription polymerase chain reaction. The signal transduction pathways were examined by Western blotting. RESULTS: None of the sealers were cytotoxic. ARS, MTA Fillapex, and iRoot SP induced a lower expression of proinflammatory mediators than Sealapex. All sealers increased ALP activity and the formation of mineralized nodules and up-regulated the expression of osteoblastic marker messenger RNA. ARS, MTA Fillapex, and iRoot SP showed superior osteogenic potential compared with Sealapex. The expression and/or activation of integrin receptors and downstream signaling molecules, including focal adhesion kinase, paxillin, Akt, mitogen-activated protein kinase, and nuclear factor κB, was induced by ARS, MTA Fillapex, and iRoot SP treatment but not by Sealapex treatment. CONCLUSIONS: We show for the first time that ARS, MTA Fillapex, and iRoot SP induce a lower expression of inflammatory mediators and enhance osteoblastic differentiation of PDLCs via the integrin-mediated signaling pathway compared with Sealapex.


Assuntos
Materiais Biocompatíveis/farmacologia , Mediadores da Inflamação/farmacologia , Osteogênese/efeitos dos fármacos , Materiais Restauradores do Canal Radicular/farmacologia , Fosfatase Alcalina/análise , Fosfatase Alcalina/efeitos dos fármacos , Compostos de Alumínio/farmacologia , Compostos de Alumínio/toxicidade , Antraquinonas , Materiais Biocompatíveis/toxicidade , Compostos de Cálcio/farmacologia , Compostos de Cálcio/toxicidade , Hidróxido de Cálcio/farmacologia , Hidróxido de Cálcio/toxicidade , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Corantes , Combinação de Medicamentos , Durapatita/farmacologia , Durapatita/toxicidade , Quinase 1 de Adesão Focal/efeitos dos fármacos , Humanos , Mediadores da Inflamação/toxicidade , Teste de Materiais , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Nanopartículas , Osteoblastos/efeitos dos fármacos , Óxidos/farmacologia , Óxidos/toxicidade , Paxilina/efeitos dos fármacos , Ligamento Periodontal/citologia , Ligamento Periodontal/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Materiais Restauradores do Canal Radicular/toxicidade , Salicilatos/farmacologia , Salicilatos/toxicidade , Transdução de Sinais/efeitos dos fármacos , Silicatos/farmacologia , Silicatos/toxicidade , Sais de Tetrazólio , Tiazóis
13.
J Endod ; 40(2): 217-22, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24461407

RESUMO

INTRODUCTION: Dental pulp is particularly susceptible to ischemic conditions (hypoxia and serum deprived) because it is commonly exposed to trauma, inflammation, chronic caries injury, and pulpitis. We investigated the apoptotic response of human dental pulp cells (HDPCs) to varying levels of oxygen and serum to mimic different degrees of ischemia, tested whether lysophosphatidic acid (LPA) could reverse ischemia-induced apoptosis, and investigated the possible mechanisms of LPA. METHODS: HDPCs were cultured under conditions mimicking serum deprivation and ischemia for 2 days with or without LPA at 25 µg/mL. Flow cytometry and JC-1 fluorescence were used to detect any apoptotic change. Western blotting was used to measure the expression of the apoptosis regulators B-cell lymphoma 2 (Bcl-2) and Bax, focal adhesion kinase (FAK), Src, extracellular signal-regulated kinase (ERK), and Akt. RESULTS: Flow cytometry and JC-1 immunofluorescence showed that ischemia could induce apoptosis of HDPCs in 2 days and treatment with LPA could reduce cell death significantly. To clarify the molecular mechanisms, Western blot results showed up-regulation of both proapoptotic Bax and antiapoptotic Bcl-2 during apoptosis. LPA functioned as an antiapoptotic cytokine by activation of the phosphorylation of FAK and ERK. No statistically significant difference was found in the activation levels of p-Src or p-Akt. CONCLUSIONS: A self-defense mechanism functioned during cell apoptosis. LPA could effectively rescue HDPCs from ischemia-induced apoptosis via regulation of Bax and Bcl-2 and the activation of phosphorylated FAK and phosphorylated ERK. LPA is a potent candidate for biological therapy of chronic pulpal inflammatory diseases.


Assuntos
Apoptose/efeitos dos fármacos , Polpa Dentária/citologia , Isquemia/fisiopatologia , Lisofosfolipídeos/farmacologia , Substâncias Protetoras/farmacologia , Adolescente , Adulto , Técnicas de Cultura de Células , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Microambiente Celular/efeitos dos fármacos , Microambiente Celular/fisiologia , Polpa Dentária/irrigação sanguínea , Polpa Dentária/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/análise , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Quinase 1 de Adesão Focal/análise , Quinase 1 de Adesão Focal/efeitos dos fármacos , Humanos , Isquemia/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/análise , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/análise , Proteínas Proto-Oncogênicas pp60(c-src)/efeitos dos fármacos , Adulto Jovem , Proteína X Associada a bcl-2/análise , Proteína X Associada a bcl-2/efeitos dos fármacos
14.
Wound Repair Regen ; 21(4): 634-40, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23815230

RESUMO

Our finding that human skin expresses leucine-rich glioma inactivated 3 (LGI3) raises the question of the function of this cytokine in keratinocytes. We have shown that LGI3 stimulates human HaCaT keratinocyte migration without affecting viability or proliferation. Western blot analysis showed that LGI3 induced focal adhesion kinase activation, Akt phosphorylation, and glycogen synthase kinase 3ß (GSK3ß) phosphorylation in these cells. Using the scratch wound assay and a modified Boyden chamber, we found that LY294002, a selective phosphatidylinositol 3-kinase inhibitor, and LiCl, a selective GSK3ß inhibitor, abolished LGI3-induced cell migration. We tested ß-catenin levels after LGI3 treatment because the Akt-GSK3ß pathway regulates ß-catenin accumulation, and ß-catenin promotes cell migration. LGI3 treatment increased ß-catenin protein and nuclear localization, whereas LY294002 prevented LGI3-induced focal adhesion kinase and Akt activation as well as ß-catenin accumulation. Overall, these data suggest that LGI3 stimulates HaCaT cell migration following ß-catenin accumulation through the Akt pathway.


Assuntos
Movimento Celular/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Proteínas/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Quinase 3 da Glicogênio Sintase/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Queratinócitos/fisiologia , Proteínas do Tecido Nervoso , Proteína Oncogênica v-akt/efeitos dos fármacos , Proteína Oncogênica v-akt/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Pele/citologia , Pele/metabolismo , beta Catenina/efeitos dos fármacos , beta Catenina/metabolismo
15.
Stem Cells ; 28(3): 555-63, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20073042

RESUMO

The integrity of transplanted mesenchymal stem cells (MSCs) for cardiac regeneration is dependent on cell-cell or cell-matrix adhesion, which is inhibited by reactive oxygen species (ROS) generated in ischemic surroundings after myocardial infarction. Intracellular ROS play a key role in the regulation of cell adhesion, migration, and proliferation. This study was designed to investigate the role of ROS on MSC adhesion. In H(2)O(2) treated MSCs, adhesion and spreading were inhibited and detachment was increased in a dose-dependent manner, and these effects were significantly rescued by co-treatment with the free radical scavenger, N-acetyl-L-cysteine (NAC, 1 mM). A similar pattern was observed on plates coated with different matrices such as fibronectin and cardiogel. Hydrogen peroxide treatment resulted in a marked decrease in the level of focal adhesion-related molecules, such as phospho-FAK and p-Src in MSCs. We also observed a significant decrease in the integrin-related adhesion molecules, alpha V and beta1, in H(2)O(2) treated MSCs. When injected into infarcted hearts, the adhesion of MSCs co-injected with NAC to the border region was significantly improved. Consequently, we observed that fibrosis and infarct size were reduced in MSC and NAC-injected rat hearts compared to in MSC-only injected hearts. These results indicate that ROS inhibit cellular adhesion of engrafted MSCs and provide evidence that the elimination of ROS might be a novel strategy for improving the survival of engrafted MSCs.


Assuntos
Moléculas de Adesão Celular/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/cirurgia , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Moléculas de Adesão Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Sequestradores de Radicais Livres/farmacologia , Técnicas de Introdução de Genes , Sobrevivência de Enxerto/fisiologia , Peróxido de Hidrogênio/farmacologia , Integrinas/efeitos dos fármacos , Integrinas/metabolismo , Masculino , Isquemia Miocárdica/fisiopatologia , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Regeneração/fisiologia , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
16.
Exp Cell Res ; 315(18): 3187-200, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19683525

RESUMO

Laminin-5 and alpha3beta1 integrin promote keratinocyte survival; however, the downstream signaling pathways for laminin-5/alpha3beta1 integrin-mediated cell survival had not been fully established. We report the unexpected finding of multiple interactions between 14-3-3 isoforms and proapoptotic proteins in the survival signaling pathway. Ln5-P4 motif within human laminin-5 alpha3 chain promotes cell survival and anti-apoptosis by inactivating Bad and YAP. This effect is achieved through the formation of 14-3-3zeta/p-Bad and 14-3-3sigma/p-YAP complexes, which is initiated by alpha3beta1 integrin and FAK/PI3K/Akt signaling. These complexes result in cytoplasmic sequestration of Bad and YAP and their subsequent inactivation. An increase in Akt1 activity in cells induces 14-3-3zeta and sigma, p-Bad, and p-YAP, promoting cell survival, whereas decreasing Akt activity suppresses the same proteins and inhibits cell survival. Suppression of 14-3-3zeta with RNA-interference inhibits cell viability and promotes apoptosis. These results reveal a new mechanism of cell survival whereby the formation of 14-3-3zeta/p-Bad and 14-3-3sigma/p-YAP complexes is initiated by laminin-5 stimulation via the alpha3beta1 integrin and FAK/PI3K/Akt signaling pathways, thereby resulting in cell survival and anti-apoptosis.


Assuntos
Proteínas 14-3-3/metabolismo , Integrina alfa3beta1/metabolismo , Queratinócitos/metabolismo , Cicatrização , Proteínas 14-3-3/agonistas , Proteínas 14-3-3/genética , Motivos de Aminoácidos/fisiologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Moléculas de Adesão Celular/farmacologia , Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Pré-Escolar , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Lactente , Integrina alfa3beta1/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Morfolinas/farmacologia , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteína de Morte Celular Associada a bcl/efeitos dos fármacos , Proteína de Morte Celular Associada a bcl/metabolismo , Calinina
17.
Cell Tissue Res ; 337(2): 243-55, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19484266

RESUMO

The related cytoplasmic non-receptor tyrosine kinases Pyk2 (proline-rich tyrosine kinase 2) and FAK (focal adhesion kinase) have been implicated in phenylephrine-induced G-protein-coupled receptor-mediated signaling mechanisms leading to cardiomyocyte hypertrophy. We report that, in phenylephrine-stimulated neonatal rat ventricular myocytes (NRVM), Pyk2 augments expression of the hypertrophic marker atrial natriuretic factor (ANF) but reduces cytoskeletal organization and cell spreading. In contrast, FAK attenuates ANF production but does not alter cytoskeletal organization and cell spreading. Pyk2 and FAK exhibit differential localization in both unstimulated and phenylephrine-stimulated myocytes. Pyk2 catalytic activity is required for Pyk2 to augment ANF secretion but is not necessary to reduce cell spreading. Pyk2 autophosphorylation is required but not sufficient for Pyk2 to augment ANF secretion. Expression of the Pyk2 FERM domain as an autonomous fragment inhibits phenylephrine-mediated ANF secretion and reduces cell spreading. In addition, expression of the Pyk2 FERM domain inhibits the ability of Pyk2 to augment ANF secretion; this is correlated with reduced Pyk2 autophosphorylation. These data indicate that Pyk2 and FAK have different roles and occupy different positions in signaling pathways leading to the development of cardiomyocyte hypertrophy.


Assuntos
Cardiomegalia/enzimologia , Citoesqueleto/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Miócitos Cardíacos/enzimologia , Animais , Fator Natriurético Atrial/agonistas , Fator Natriurético Atrial/biossíntese , Miosinas Cardíacas/metabolismo , Cardiomegalia/patologia , Cardiotônicos/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 2 de Adesão Focal/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Cadeias Leves de Miosina/metabolismo , Fenilefrina/farmacologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transfecção
18.
Neurosci Res ; 64(4): 348-54, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19379779

RESUMO

Overexpression of angiopoietin (Ang) 1 in the brain results in increased vascularization and altered neuronal dendrite configuration. We hypothesized that Ang1 acts directly on neurons inducing neurite outgrowth. We stimulated PC12 cells with Ang1 and observed outgrowth levels comparable to nerve growth factor (NGF). Western blotting and RT-PCR demonstrated the absence of the Ang1 receptor, Tie2 and the presence of beta1-integrin. Downstream of beta1-integrin, Ang1 stimulation led to a approximately 2.6 fold increase in focal adhesion kinase (FAK) phosphorylation and no change in the activation of mitogen-activated protein kinase (MAPK) nor c-Jun N-terminal kinase (JNK). Conversely, NGF stimulation had no effect on FAK phosphorylation but led to a approximately 3.1 and approximately 2 fold increase in phosphorylation of MAPK and JNK. Ang1, but not NGF-mediated outgrowth was attenuated following functional inhibition of beta1-integrin and FAK, and Wortmannin inhibited neurite outgrowth mediated by both. Our results suggest that Ang1 induces neurite outgrowth in PC12 cells in a Tie2-independent, beta1-integrin-FAK-PI3K-Akt-dependent manner and that NGF and Ang1 mediate neurite outgrowth via two independent signaling mechanisms.


Assuntos
Angiopoietina-1/análogos & derivados , Diferenciação Celular/fisiologia , Cadeias beta de Integrinas/metabolismo , Neuritos/metabolismo , Receptor TIE-2/metabolismo , Transdução de Sinais/fisiologia , Angiopoietina-1/metabolismo , Angiopoietina-1/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Cadeias beta de Integrinas/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Células PC12 , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor TIE-2/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
19.
Front Biosci ; 13: 6604-16, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18508682

RESUMO

A monomeric RGD-disintegrin was recently identified from a cDNA library from the venom gland of Bothrops alternatus. The corresponding 12 kDa-recombinant protein, DisBa-01, specifically interacted with alpha(v)beta3 integrin and displayed potent anti-metastatic and anti-angiogenic properties. Here, the interaction of DisBa-01 with platelet alphaIIb beta3 integrin and its effects on hemostasis and thrombosis were investigated. DisBa-01 bound to Chinese Hamster Ovary (CHO) cells expressing beta3 or alphaIIb beta3 and promoted their adhesion and the adhesion of resting platelets onto glass coverslips. The disintegrin inhibited the binding of FITC-fibrinogen and FITC-PAC-1 to ADP-stimulated platelets and inhibited ADP-, TRAP- and collagen-induced aggregation of murine, rabbit or human platelets. In a flow chamber assay, DisBa-01 inhibited and reverted platelet adhesion to immobilized fibrinogen. DisBa-01 inhibited the phosphorylation of FAK following platelet activation. The intravenous injection of DisBa-01 in C57Bl6/j mice, prolonged tail bleeding time as well as thrombotic occlusion time in mesenteric venules and arterioles following vessel injury with FeCl3. In conclusion, DisBa-01 antagonizes the platelet alphaIIb beta3 integrin and potently inhibits thrombosis.


Assuntos
Venenos de Crotalídeos/toxicidade , Hemostasia/efeitos dos fármacos , Hemostáticos/farmacologia , Animais , Bothrops , Células CHO/efeitos dos fármacos , Cricetinae , Cricetulus , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Humanos , Fosforilação , Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Proteínas Recombinantes/toxicidade , Transfecção
20.
Carcinogenesis ; 29(6): 1096-107, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18263593

RESUMO

Pancreatic cancer is a lethal disease accounting for the fourth leading cause of cancer death in USA. Focal adhesion kinase (FAK) and the insulin-like growth factor-I receptor (IGF-1R) are tyrosine kinases that activate common pathways, leading to increased proliferation and cell survival. Sparse information is available regarding their contribution to the malignant behavior of pancreatic cancer. We analyzed the relationship between FAK and IGF-1R in human pancreatic cancer cells, determined which downstream signaling pathways are altered following kinase inhibition or downregulation and studied whether dual kinase inhibition represents a potential novel treatment strategy in this deadly disease. Using immunoprecipitation and confocal microscopy, we show for the first time that FAK and IGF-1R physically interact in pancreatic cancer cells and that inhibition of tyrosine phosphorylation of either kinase disrupts their interaction. Decreasing phosphorylation of either FAK or IGF-1R alone resulted in little inhibition of cell viability or increased apoptosis. However, dual inhibition of FAK, using either a dominant-negative construct (FAK-CD) or small interfering RNA, and IGF-1R, using a specific small molecule tyrosine kinase inhibitor (AEW-541) or stable expression of a truncated, mutated IGF-1R, led to a synergistic decrease in cell proliferation and phosphorylation of extracellular signal-regulated kinase (ERK) and increase in cell detachment and apoptosis compared with inhibition of either pathway alone. Dual kinase inhibition with FAK-CD and AEW-541 resulted in a marked increase in apoptosis when FAK was displaced from the focal adhesions. Inhibition of both tyrosine kinase activities via a novel single small molecular inhibitor (TAE 226), at low doses specific for FAK and IGF-1R, resulted in significant inhibition of cell viability, decrease in phosphorylation of ERK and Akt and increase in apoptosis accompanied by cleavage of Poly (ADP-ribose) polymerase (PARP) and activation of caspase-3 in pancreatic cancer cells. Thus, simultaneous inhibition of both tyrosine kinases represents a potential novel therapeutic approach in human pancreatic adenocarcinoma.


Assuntos
Adenocarcinoma/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Western Blotting , Linhagem Celular Tumoral , Imunofluorescência , Quinase 1 de Adesão Focal/efeitos dos fármacos , Humanos , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Camundongos , Microscopia Confocal , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno , Receptor IGF Tipo 1/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA