Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pak J Pharm Sci ; 34(3): 891-898, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34602411

RESUMEN

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.


Asunto(s)
Alcaloides/farmacología , Carbolinas/farmacología , Carcinoma/metabolismo , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Peganum , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/metabolismo , Serina-Treonina Quinasas TOR/efectos de los fármacos , Animales , Carcinoma/genética , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Ratones , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Gástricas/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
2.
Invest New Drugs ; 39(2): 377-385, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32989643

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Stichopus , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Integrina beta1/efectos de los fármacos , Neoplasias Hepáticas/patología , Masculino , Toxinas Marinas/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/prevención & control , Péptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Cell Physiol ; 235(11): 8334-8344, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32239671

RESUMEN

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.


Asunto(s)
Uniones Adherentes/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Quinasa 1 de Adhesión Focal/metabolismo , Metformina/farmacología , Uniones Adherentes/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Humanos , Transporte de Proteínas/efectos de los fármacos
4.
J Med Chem ; 62(5): 2508-2520, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30739444

RESUMEN

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.


Asunto(s)
Quinasa 1 de Adhesión Focal/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes/metabolismo , Línea Celular Tumoral , Proliferación Celular , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Ligandos , Proteolisis , Interferencia de ARN
5.
Life Sci ; 213: 248-257, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30292831

RESUMEN

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.


Asunto(s)
Dasatinib/farmacología , Rosuvastatina Cálcica/farmacología , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Masculino , Ratones , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/efectos de los fármacos
6.
Oncotarget ; 8(13): 21674-21691, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28423510

RESUMEN

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.


Asunto(s)
Anticarcinógenos/farmacología , Genisteína/farmacología , Melanoma Experimental/patología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Etiquetado Corte-Fin in Situ , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Invasividad Neoplásica/patología , Paxillin/efectos de los fármacos , Paxillin/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
World J Urol ; 35(8): 1247-1254, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28013345

RESUMEN

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.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Quinasa 1 de Adhesión Focal/genética , Integrina alfa4/genética , Integrina alfaV/genética , Sistema de Señalización de MAP Quinasas/genética , Miocitos del Músculo Liso/metabolismo , Fenómenos Biomecánicos , Western Blotting , Colágeno Tipo I/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo III/efectos de los fármacos , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Proteínas de la Matriz Extracelular/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas/efectos de los fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Flavonoides/farmacología , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Integrina alfa4/efectos de los fármacos , Integrina alfa4/metabolismo , Integrina alfaV/efectos de los fármacos , Integrina alfaV/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Vejiga Urinaria/citología
8.
Eur J Oral Sci ; 123(4): 249-53, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26058789

RESUMEN

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.


Asunto(s)
Fibroblastos/efectos de los fármacos , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Interleucina-6/análisis , Interleucina-8/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Ligamento Periodontal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Fibroblastos/enzimología , Quinasa 1 de Adhesión Focal/genética , Técnicas de Silenciamiento del Gen , Humanos , Ligamento Periodontal/citología , Ligamento Periodontal/enzimología , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/administración & dosificación
9.
J Dent Res ; 94(6): 853-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25724555

RESUMEN

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.


Asunto(s)
Compuestos de Calcio/farmacología , Cerámica/química , Pulpa Dental/citología , Materiales de Recubrimiento Pulpar y Pulpectomía/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Silicatos/farmacología , Compuestos de Aluminio/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Pulpa Dental/efectos de los fármacos , Dentina Secundaria/efectos de los fármacos , Dentinogénesis/efectos de los fármacos , Combinación de Medicamentos , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ensayo de Materiales , Proteínas de la Membrana/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Nanopartículas/química , Óxidos/farmacología , Paxillin/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Ratas , Fibras de Estrés/efectos de los fármacos , Vinculina/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
10.
Cancer Prev Res (Phila) ; 8(5): 419-30, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25712051

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas/patología , Matriz Extracelular/efectos de los fármacos , Fenretinida/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Neoplasias de la Boca/patología , Lesiones Precancerosas/patología , Anticarcinógenos/farmacología , Movimiento Celular/efectos de los fármacos , Quimioprevención , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/patología , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
11.
J Endod ; 40(10): 1642-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25260738

RESUMEN

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.


Asunto(s)
Materiales Biocompatibles/farmacología , Mediadores de Inflamación/farmacología , Osteogénesis/efectos de los fármacos , Materiales de Obturación del Conducto Radicular/farmacología , Fosfatasa Alcalina/análisis , Fosfatasa Alcalina/efectos de los fármacos , Compuestos de Aluminio/farmacología , Compuestos de Aluminio/toxicidad , Antraquinonas , Materiales Biocompatibles/toxicidad , Compuestos de Calcio/farmacología , Compuestos de Calcio/toxicidad , Hidróxido de Calcio/farmacología , Hidróxido de Calcio/toxicidad , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colorantes , Combinación de Medicamentos , Durapatita/farmacología , Durapatita/toxicidad , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Humanos , Mediadores de Inflamación/toxicidad , Ensayo de Materiales , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Nanopartículas , Osteoblastos/efectos de los fármacos , Óxidos/farmacología , Óxidos/toxicidad , Paxillin/efectos de los fármacos , Ligamento Periodontal/citología , Ligamento Periodontal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Materiales de Obturación del Conducto Radicular/toxicidad , Salicilatos/farmacología , Salicilatos/toxicidad , Transducción de Señal/efectos de los fármacos , Silicatos/farmacología , Silicatos/toxicidad , Sales de Tetrazolio , Tiazoles
12.
J Endod ; 40(2): 217-22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24461407

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Pulpa Dental/citología , Isquemia/fisiopatología , Lisofosfolípidos/farmacología , Sustancias Protectoras/farmacología , Adolescente , Adulto , Técnicas de Cultivo de Célula , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Microambiente Celular/fisiología , Pulpa Dental/irrigación sanguínea , Pulpa Dental/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/análisis , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/análisis , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Humanos , Isquemia/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/análisis , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas pp60(c-src)/análisis , Proteínas Proto-Oncogénicas pp60(c-src)/efectos de los fármacos , Adulto Joven , Proteína X Asociada a bcl-2/análisis , Proteína X Asociada a bcl-2/efectos de los fármacos
13.
Wound Repair Regen ; 21(4): 634-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23815230

RESUMEN

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.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Proteínas/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Queratinocitos/fisiología , Proteínas del Tejido Nervioso , Proteína Oncogénica v-akt/efectos de los fármacos , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Piel/citología , Piel/metabolismo , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo
14.
Stem Cells ; 28(3): 555-63, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20073042

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/cirugía , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Depuradores de Radicales Libres/farmacología , Técnicas de Sustitución del Gen , Supervivencia de Injerto/fisiología , Peróxido de Hidrógeno/farmacología , Integrinas/efectos de los fármacos , Integrinas/metabolismo , Masculino , Isquemia Miocárdica/fisiopatología , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Regeneración/fisiología , Familia-src Quinasas/efectos de los fármacos , Familia-src Quinasas/metabolismo
15.
Exp Cell Res ; 315(18): 3187-200, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19683525

RESUMEN

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.


Asunto(s)
Proteínas 14-3-3/metabolismo , Integrina alfa3beta1/metabolismo , Queratinocitos/metabolismo , Cicatrización de Heridas , Proteínas 14-3-3/agonistas , Proteínas 14-3-3/genética , Secuencias de Aminoácidos/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/farmacología , Proteínas de Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Preescolar , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Lactante , Integrina alfa3beta1/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Morfolinas/farmacología , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Proteína Letal Asociada a bcl/efectos de los fármacos , Proteína Letal Asociada a bcl/metabolismo , Kalinina
16.
Cell Tissue Res ; 337(2): 243-55, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19484266

RESUMEN

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.


Asunto(s)
Cardiomegalia/enzimología , Citoesqueleto/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Miocitos Cardíacos/enzimología , Animales , Factor Natriurético Atrial/agonistas , Factor Natriurético Atrial/biosíntesis , Miosinas Cardíacas/metabolismo , Cardiomegalia/patología , Cardiotónicos/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 2 de Adhesión Focal/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Cadenas Ligeras de Miosina/metabolismo , Fenilefrina/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transfección
17.
Neurosci Res ; 64(4): 348-54, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19379779

RESUMEN

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.


Asunto(s)
Angiopoyetina 1/análogos & derivados , Diferenciación Celular/fisiología , Cadenas beta de Integrinas/metabolismo , Neuritas/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal/fisiología , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Cadenas beta de Integrinas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Células PC12 , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptor TIE-2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
Front Biosci ; 13: 6604-16, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508682

RESUMEN

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.


Asunto(s)
Venenos de Crotálidos/toxicidad , Hemostasis/efectos de los fármacos , Hemostáticos/farmacología , Animales , Bothrops , Células CHO/efectos de los fármacos , Cricetinae , Cricetulus , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Fosforilación , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Proteínas Recombinantes/toxicidad , Transfección
19.
Carcinogenesis ; 29(6): 1096-107, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18263593

RESUMEN

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.


Asunto(s)
Adenocarcinoma/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Humanos , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Ratones , Microscopía Confocal , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño , Receptor IGF Tipo 1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transfección
20.
J Cell Biochem ; 99(1): 88-95, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16598791

RESUMEN

Spreading of SNU16mAd gastric carcinoma cells was previously shown to be regulated via a signaling network from transforming growth factor beta1 (TGFbeta1) to integrins signaling, through a mediation of protein kinase C delta (PKCdelta). However, in the previous study, the roles of PKCdelta appeared complicated. In this study to clarify the roles of PKCdelta in the spreading of the gastric carcinoma cells, we questioned if PKC activation via phorbol 12-myristate 13-acetate (PMA) treatment could mimic the TGFbeta1 effects. An acute PMA treatment increased phosphorylations of focal adhesion (FA) kinase, paxillin, c-Src, and cofilin, just as TGFbeta1 did. Furthermore, cell spreading mediated by TGFbeta1- or acute PMA treatment correlated with activation of RhoA, which regulates actin reorganization and FA formation. However, stress fiber formation was prominent in TGFbeta1-treated cells, compared to cortical actin organization in PMA-treated cells. Altogether, these observations indicate that acute PMA treatment could mimic the TGFbeta1 mechanisms for cell spreading through subtly different effects on actin reorganization.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Integrinas/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Proteína Tirosina Quinasa CSK , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Cofilina 1/efectos de los fármacos , Cofilina 1/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Integrinas/efectos de los fármacos , Imitación Molecular , Paxillin/efectos de los fármacos , Paxillin/metabolismo , Fosforilación , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1 , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...