Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioorg Med Chem Lett ; 40: 127965, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33744442

RESUMEN

Small molecule inhibitors of the focal adhesion kinase are regarded as promising tools in our armamentarium for treating cancer. Here, we identified four 1,2,4-triazole derivatives that inhibit FAK kinase significantly and evaluated their therapeutic potential. Most tested compounds revealed potent antiproliferative activity in HepG2 and Hep3B liver cancer cells, in which 3c and 3d were the most potent (IC50 range; 2.88 ~ 4.83 µM). Compound 3d possessed significant FAK inhibitory activity with IC50 value of 18.10 nM better than the reference GSK-2256098 (IC50 = 22.14 nM). The preliminary mechanism investigation by Western blot analysis showed that both 3c and 3d repressed FAK phosphorylation comparable to GSK-2256098 in HepG2 cells. As a result of FAK inhibition, 3c and 3d inhibited the pro-survival pathways by decreasing the phosphorylation levels of PI3K, Akt, JNK, and STAT3 proteins. This effect led to apoptosis induction and cell cycle arrest. Taken together, these results indicate that 3d could serve as a potent preclinical candidate for the treatment of cancers.


Asunto(s)
Acetanilidas/farmacología , Aminobenzoatos/farmacología , Antineoplásicos/farmacología , Quinasa 1 de Adhesión Focal/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Triazoles/farmacología , Acetanilidas/síntesis química , Aminobenzoatos/síntesis química , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Quinasa 1 de Adhesión Focal/química , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Triazoles/síntesis química
2.
J Biol Chem ; 295(41): 14111-14124, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32753481

RESUMEN

The multifaceted adaptor protein ß-arr1 (ß-arrestin1) promotes activation of focal adhesion kinase (FAK) by the chemokine receptor CXCR4, facilitating chemotaxis. This function of ß-arr1 requires the assistance of the adaptor protein STAM1 (signal-transducing adaptor molecule 1) because disruption of the interaction between STAM1 and ß-arr1 reduces CXCR4-mediated activation of FAK and chemotaxis. To begin to understand the mechanism by which ß-arr1 together with STAM1 activates FAK, we used site-directed spin-labeling EPR spectroscopy-based studies coupled with bioluminescence resonance energy transfer-based cellular studies to show that STAM1 is recruited to activated ß-arr1 by binding to a novel surface on ß-arr1 at the base of the finger loop, at a site that is distinct from the receptor-binding site. Expression of a STAM1-deficient binding ß-arr1 mutant that is still able to bind to CXCR4 significantly reduced CXCL12-induced activation of FAK but had no impact on ERK-1/2 activation. We provide evidence of a novel surface at the base of the finger loop that dictates non-GPCR interactions specifying ß-arrestin-dependent signaling by a GPCR. This surface might represent a previously unidentified switch region that engages with effector molecules to drive ß-arrestin signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Complejos de Clasificación Endosomal Requeridos para el Transporte , Sistema de Señalización de MAP Quinasas , Fosfoproteínas , Receptores CXCR4 , beta-Arrestina 1 , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Células HEK293 , Humanos , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Estructura Secundaria de Proteína , Receptores CXCR4/química , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , beta-Arrestina 1/química , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo
3.
Molecules ; 25(15)2020 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-32751931

RESUMEN

Focal adhesion kinase (FAK) is a tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. In cancer cells, FAK promotes the progression and metastasis of tumours. In this study, we used structure-based virtual screening to filter a library of more than 210K compounds against the focal adhesion targeting FAK-focal adhesion targeting (FAT) domain to identify 25 virtual hit compounds which were screened in the invasive breast cancer line (MDA-MB-231). Most notably, compound I showed low micromolar antiproliferative activity, as well as antimigratory activity. Moreover, examination in a model of triple negative breast cancer (TNBC), revealed that, despite not effecting FAK phosphorylation, compound I significantly impairs proliferation whilst impairing focal adhesion growth and turnover leading to reduced migration. Further optimisation and synthesis of analogues of the lead compound I using a four-step synthetic procedure was performed, and analogues were assessed for their antiproliferative activity against three breast cancer (MDA-MB-231, T47D, BT474) cell lines and one pancreatic cancer (MIAPaCa2) cell line. Compound 5f was identified as a promising lead compound with IC50 values in the range of 4.59-5.28 µM in MDA-MB-231, T47D, BT474, and MIAPaCa2. Molecular modelling and pharmacokinetic studies provided more insight into the therapeutic features of this new series.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/química , Neoplasias Pancreáticas/metabolismo , Dominios Proteicos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cricetulus , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Enlace de Hidrógeno , Simulación del Acoplamiento Molecular , Neoplasias Pancreáticas/patología , Regulador Transcripcional ERG/genética , Transfección , Neoplasias de la Mama Triple Negativas/patología
4.
Biochim Biophys Acta Gen Subj ; 1864(1): 129450, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676296

RESUMEN

BACKGROUND: Leucine rich Aspartate motifs (LD motifs) are molecular recognition motifs on Paxillin that recognize LD-motif binding domains (LDBD) of a number of focal adhesion proteins in order to carry out downstream signaling and actin cytoskeleton remodeling. In this study, we identified structural features within LDBDs that influence their binding affinity with Paxillin LD motifs. METHODS: Various point mutants of focal adhesion targeting (FAT) domain of Focal Adhesion Kinase (FAK) were created by moving a key Lysine residue two and three helical turns in order to match the unique conformations as observed in LDBDs of two other focal adhesion proteins, Vinculin and CCM3. RESULTS: This led to identify a mutant of FAT domain of FAK, named as FAT(NV) (Asn992 of FAT domain was replaced by Val), with remarkable high affinity for LD1 (Kd = 1.5 µM vs no-binding with wild type) and LD2 peptides (Kd = 7.2 µM vs 63 µM with wild type). Consistently, the focal adhesions of MCF7 cells expressing FAK(NV) were highly stable (turnover rate = 1.25 × 10-5 µm2/s) as compared to wild type FAK transfected cells (turnover rate = 1.5 × 10-3 µm2/s). CONCLUSIONS: We observed that the relative disposition of key LD binding amino-acids at LDBD surface, hydrophobic burial of long Leucine side chains of LD-motifs and complementarity of charged surfaces are the key factors determining the binding affinities of LD motifs with LDBDs. GENERAL SIGNIFICANCE: Our study will help in protein engineering of FAT domain of FAK by modulating FAK-LD motif interactions which have implications in cellular focal adhesions and cell migration.


Asunto(s)
Adhesión Celular/genética , Quinasa 1 de Adhesión Focal/genética , Adhesiones Focales/genética , Conformación Proteica , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos/genética , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Ácido Aspártico/genética , Sitios de Unión/genética , Movimiento Celular/genética , Quinasa 1 de Adhesión Focal/química , Adhesiones Focales/química , Regulación de la Expresión Génica/genética , Humanos , Lisina/química , Lisina/genética , Células MCF-7 , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Paxillin/química , Paxillin/genética , Unión Proteica/genética , Ingeniería de Proteínas , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Vinculina/química , Vinculina/genética
5.
SLAS Discov ; 25(1): 21-32, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31513463

RESUMEN

Focal adhesion kinase (FAK) is a promising cancer drug target due to its massive overexpression in multiple solid tumors and its critical role in the integration of signals that control proliferation, invasion, apoptosis, and metastasis. Previous FAK drug discovery and high-throughput screening have exclusively focused on the identification of inhibitors that target the kinase domain of FAK. Because FAK is both a kinase and scaffolding protein, the development of novel screening assays that detect inhibitors of FAK protein-protein interactions remains a critical need. In this report, we describe the development of a high-throughput fluorescence polarization (FP) screening assay that measures the interactions between FAK and paxillin, a focal adhesion-associated protein. We designed a tetramethylrhodamine (TAMRA)-tagged paxillin peptide based on the paxillin LD2 motif that binds to the focal adhesion targeting (FAT) domain with significant dynamic range, specificity, variability, stability, and a Z'-factor suitable for high-throughput screening. In addition, we performed a pilot screen of 1593 compounds using this FP assay, showing its feasibility for high-throughput drug screening. Finally, we identified three compounds that show dose-dependent competition of FAT-paxillin binding. This assay represents the first described high-throughput screening assay for FAK scaffold inhibitors and can accelerate drug discovery efforts for this promising drug target.


Asunto(s)
Descubrimiento de Drogas , Polarización de Fluorescencia , Quinasa 1 de Adhesión Focal/metabolismo , Ensayos Analíticos de Alto Rendimiento , Paxillin/metabolismo , Unión Proteica/efectos de los fármacos , Descubrimiento de Drogas/métodos , Polarización de Fluorescencia/métodos , Quinasa 1 de Adhesión Focal/química , Humanos , Modelos Moleculares , Conformación Molecular , Paxillin/química , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad
6.
Cell Death Dis ; 10(10): 770, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601793

RESUMEN

Ovarian cancer remains the most lethal gynecologic malignancy with late detection and acquired chemoresistance. Advanced understanding of the pathophysiology and novel treatment strategies are urgently required. A growing body of proteomic investigations suggest that phosphorylation has a pivotal role in the regulation of ovarian cancer associated signaling pathways. Matrine has been extensively studied for its potent anti-tumor activities. However, its effect on ovarian cancer cells and underlying molecular mechanisms remain unclear. Herein we showed that matrine treatment inhibited the development and progression of ovarian cancer cells by regulating proliferation, apoptosis, autophagy, invasion and angiogenesis. Matrine treatment retarded the cancer associated signaling transduction by decreasing the phosphorylation levels of ERK1/2, MEK1/2, PI3K, Akt, mTOR, FAK, RhoA, VEGFR2, and Tie2 in vitro and in vivo. Moreover, matrine showed excellent antitumor effect on chemoresistant ovarian cancer cells. No obvious toxic side effects were observed in matrine-administrated mice. As the natural agent, matrine has the potential to be the targeting drug against ovarian cancer cells with the advantages of overcoming the chemotherapy resistance and decreasing the toxic side effects.


Asunto(s)
Alcaloides/uso terapéutico , Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Quinolizinas/uso terapéutico , Alcaloides/efectos adversos , Alcaloides/farmacología , Animales , Apoptosis/efectos de los fármacos , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolizinas/efectos adversos , Quinolizinas/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/química , Serina-Treonina Quinasas TOR/metabolismo , Trasplante Heterólogo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/química , Proteína de Unión al GTP rhoA/metabolismo , Matrinas
7.
Molecules ; 24(18)2019 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-31540099

RESUMEN

The Focal Adhesion Targeting (FAT) domain of Focal Adhesion Kinase (FAK) is a promising drug target since FAK is overexpressed in many malignancies and promotes cancer cell metastasis. The FAT domain serves as a scaffolding protein, and its interaction with the protein paxillin localizes FAK to focal adhesions. Various studies have highlighted the importance of FAT-paxillin binding in tumor growth, cell invasion, and metastasis. Targeting this interaction through high-throughput screening (HTS) provides a challenge due to the large and complex binding interface. In this report, we describe a novel approach to targeting FAT through fragment-based drug discovery (FBDD). We developed two fragment-based screening assays-a primary SPR assay and a secondary heteronuclear single quantum coherence nuclear magnetic resonance (HSQC-NMR) assay. For SPR, we designed an AviTag construct, optimized SPR buffer conditions, and created mutant controls. For NMR, resonance backbone assignments of the human FAT domain were obtained for the HSQC assay. A 189-compound fragment library from Enamine was screened through our primary SPR assay to demonstrate the feasibility of a FAT-FBDD pipeline, with 19 initial hit compounds. A final total of 11 validated hits were identified after secondary screening on NMR. This screening pipeline is the first FBDD screen of the FAT domain reported and represents a valid method for further drug discovery efforts on this difficult target.


Asunto(s)
Quinasa 1 de Adhesión Focal/química , Adhesiones Focales/química , Resonancia Magnética Nuclear Biomolecular , Resonancia por Plasmón de Superficie , Descubrimiento de Drogas , Quinasa 1 de Adhesión Focal/genética , Adhesiones Focales/genética , Humanos , Paxillin/química , Paxillin/genética , Dominios Proteicos
8.
Mol Cancer Ther ; 18(1): 139-146, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30373932

RESUMEN

TRAF2, a RING finger adaptor protein, plays an important function in tumor necrosis factor (TNF)- and TNF-like weak inducer of apoptosis (TWEAK)-dependent signaling, in particular during inflammatory and immune responses. We identified a functional interaction of TRAF2 with focal adhesion (FA) signaling involving the focal adhesion kinase (FAK) in the regulation of cell susceptibility to anoikis. Comparison of TRAF2-proficient (TRAF2+/+) versus TRAF2-deficient (TRAF2-/-), and FAK-proficient (FAK+/+) versus FAK-deficient (FAK-/-) mouse embryonic fibroblasts and their matched reconstituted cells demonstrated that TRAF2 interacts physically with the N-terminal portion of FAK and colocalizes to cell membrane protrusions. This interaction was found to be critical for promoting resistance to cell anoikis. Similar results were confirmed in the human breast cancer cell line MDA-MB-231, where TRAF2 and FAK downregulation promoted cell susceptibility to anoikis. In human breast cancer tissues, genomic analysis of The Cancer Genome Atlas database revealed coamplification of TRAF2 and FAK in breast cancer tissues with a predictive value for shorter survival, further supporting a potential role of TRAF2-FAK cooperative signaling in cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/metabolismo , Animales , Anoicis , Sitios de Unión , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Regulación hacia Abajo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Quinasa 1 de Adhesión Focal/química , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones , Transducción de Señal , Análisis de Supervivencia
9.
Bioorg Med Chem ; 27(2): 305-314, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30554970

RESUMEN

A series of novel metronidazole aryloxy, carboxy and azole derivatives has been synthesized and their cytotoxic activities on three cancer cell lines were evaluated by MTT assay. Compounds 4m, 4l and 4d showed the most potent cytotoxic activity (IC50s less than 100 µg/mL). Apoptosis was also detected for these compounds by flow cytometry. Docking studies were performed in order to propose the probable target protein. In the next step, molecular dynamics simulation was carried out on the proposed target protein, focal adhesion kinase (FAK, PDB code: 2ETM), bound to compound 4m. As, 4m showed a potent cytotoxic activity and an acceptable apoptotic effect, it can be a potential anticancer candidate that may work through inhibition of FAK.


Asunto(s)
Antineoplásicos/farmacología , Metronidazol/análogos & derivados , Metronidazol/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Enlace de Hidrógeno , Metronidazol/síntesis química , Metronidazol/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad Cuantitativa
10.
J Labelled Comp Radiopharm ; 61(14): 1058-1068, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30193401

RESUMEN

A newly synthesized s-triazine derivative 1,1',1″-(((1,3,5-triazine-2,4,6-triyl) tris (azanediyl)) tris (benzene-4,1-diyl))tris (ethan-1-one), (1), was synthesized as a part of an ongoing research for development of novel s-triazine-based radiopharmaceuticals. In-vitro cell viability assay against different human cancer cell lines showed very promising inhibitory activity of the synthesized compound. This finding encouraged the radioiodination of 1 to study the degree of its localization in tumor site for evaluating the possibility of its use as a tumor imaging agent. The biodistribution study showed good localization of the radioiodinated derivative 2 at tumor site following i.v. administration in solid tumor-bearing mice. Finally, in a trial to understand the mechanism of the anticancer effect exerted by 1, a target prediction study and a docking study were performed. The results of the first study showed that focal adhesion kinase is a possible target for compound 1 and the docking study confirmed successful binding of both compound 1 and its radioiodinated derivative 2 to the binding site of focal adhesion kinase. As a conclusion, the results of this study suggest that, compound 2 could be used as a potential agent for tumor imaging after preclinical trials.


Asunto(s)
Radioisótopos de Yodo/química , Triazinas/química , Triazinas/farmacocinética , Animales , Transporte Biológico , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Conformación Proteica , Radioquímica , Distribución Tisular , Triazinas/metabolismo
11.
Neurol Sci ; 39(8): 1361-1374, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29789968

RESUMEN

Cas scaffolding protein family member 4 and protein tyrosine kinase 2 are signaling proteins, which are involved in neuritic plaques burden, neurofibrillary tangles, and disruption of synaptic connections in Alzheimer's disease. In the current study, a computational approach was employed to explore the active binding sites of Cas scaffolding protein family member 4 and protein tyrosine kinase 2 proteins and their significant role in the activation of downstream signaling pathways. Sequential and structural analyses were performed on Cas scaffolding protein family member 4 and protein tyrosine kinase 2 to identify their core active binding sites. Molecular docking servers were used to predict the common interacting residues in both Cas scaffolding protein family member 4 and protein tyrosine kinase 2 and their involvement in Alzheimer's disease-mediated pathways. Furthermore, the results from molecular dynamic simulation experiment show the stability of targeted proteins. In addition, the generated root mean square deviations and fluctuations, solvent-accessible surface area, and gyration graphs also depict their backbone stability and compactness, respectively. A better understanding of CAS and their interconnected protein signaling cascade may help provide a treatment for Alzheimer's disease. Further, Cas scaffolding protein family member 4 could be used as a novel target for the treatment of Alzheimer's disease by inhibiting the protein tyrosine kinase 2 pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Simulación del Acoplamiento Molecular , Dinámicas no Lineales , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Sitios de Unión , Femenino , Quinasa 1 de Adhesión Focal/química , Humanos , Masculino , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal
12.
Int J Biochem Cell Biol ; 95: 73-84, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29274473

RESUMEN

Tyro3, a member of TAM receptor tyrosine kinase family, has been implicated in the regulation of melanoma progression and survival. In this study, we sought the molecular mechanism of Tyro3 effects avoiding endogenous background by overexpression of Tyro3 in fibroblasts that have negligible levels of Tyro3. This introduction triggers the tyrosyl-phosphorylation of ACTN4, a member of actin binding protein family involved in motility, a behavior critical for invasive progression, as shown by siRNA to Tyro3 limiting melanoma cell migration and invasion. Tyro3-mediated phosphorylation of ACTN4 required FAK activation at tyrosine 397 and the EGF receptor cascade, but not EGFR ligand binding. Using PCR-based mutagenesis, the sites of Tyro3-mediated ACTN4 phosphorylation were mapped to ACTN4 tyrosine 11 and 13, and this occurs in conjunction with EGF-mediated phosphorylation on Y4 and Y31. Interestingly, Tyro3-mediated phosphorylation only slightly decreases the actin binding activity of ACTN4. However, this rendered the phosphorylated ACTN4 resistant to the m-calpain cleavage between Y13 and G14, a limited proteolysis that prevents growth factor regulation of ACTN4 interaction with F-actin. Overexpression of both WT ACTN4 and ACTN4Y11/13E, a mimic of ACTN4 phosphorylated at tyrosine 11 and 13, in melanoma WM983b cells resulted in a likely mesenchymal to amoeboidal transition. ACTN4Y11/13E-expressing cells were more amoeboidal, less migratory on collagen I gel coated surface but more invasive through collagen networks. In parallel, expression of ACTN4Y11/13E, in ACTN4 knockdown melanoma WM1158 cells resulted in an increase of invasion compared to WT ACTN4. These findings suggest that Tyro3-mediated phosphorylation of ACTN4 is involved in invasion of melanoma cells.


Asunto(s)
Actinina/metabolismo , Calpaína/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Melanoma/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Actinina/química , Actinina/genética , Sustitución de Aminoácidos , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Activación Enzimática , Quinasa 1 de Adhesión Focal/química , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Melanoma/enzimología , Melanoma/patología , Mutación , Invasividad Neoplásica/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilación , Estabilidad Proteica , Proteolisis , Interferencia de ARN , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Tirosina/metabolismo
13.
J Biol Chem ; 292(39): 16321-16332, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28821612

RESUMEN

The syndecan family of heparan sulfate proteoglycans contributes to cell adhesion and communication by serving as co-receptors for cell signaling and extracellular matrix molecules. Syndecan-2 is located at the cell surface, and we previously reported that it induces matrix metalloproteinase-7 (MMP-7) expression in colon cancer cells. However, the underlying regulatory mechanisms are unknown. Here, we report that overexpression of syndecan-2 in HT-29 colon cancer cells increases the phosphorylation of focal adhesion kinase (FAK) and ERK in parallel with up-regulated MMP-7 expression, but a syndecan-2 mutant lacking the cytoplasmic domain showed significant reductions in these effects. Consistent with this observation, FAK inhibition via FAK-related non-kinase expression or inhibition of ERK with the ERK1/2 inhibitor SCH772984 diminished the syndecan-2-mediated up-regulation of MMP-7. Activation of PKC enhanced syndecan-2-mediated MMP-7 expression, whereas inhibition of PKC had the opposite effect. Of note, the exogenous expression of syndecan-2 triggered localization of PKCγ to the membrane. Expression of syndecan-2 harboring a phosphomimetic (S198E) mutation of the variable region of the cytoplasmic domain enhanced MMP-7 expression and FAK phosphorylation. Finally, experimental suppression of shedding of the syndecan-2 extracellular domain did not significantly affect the syndecan-2-mediated up-regulation of MMP-7 in the early period after syndecan-2 overexpression. Taken together, these findings suggest that syndecan-2's cytoplasmic domain up-regulates MMP-7 expression in colon cancer cells via PKCγ-mediated activation of FAK/ERK signaling.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa C/metabolismo , Procesamiento Proteico-Postraduccional , Sindecano-2/metabolismo , Sustitución de Aminoácidos , Animales , Carcinoma/tratamiento farmacológico , Carcinoma/enzimología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/enzimología , Inducción Enzimática/efectos de los fármacos , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sindecano-2/antagonistas & inhibidores , Sindecano-2/química , Sindecano-2/genética
14.
J Biomol NMR ; 68(4): 237-247, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28711957

RESUMEN

An improved expression protocol is proposed for amino acid type-specific [13C], [15N]-isotope labeling of proteins in baculovirus-infected (BV) insect cell cultures. This new protocol modifies the methods published by Gossert et al. (J Biomol NMR 51(4):449-456, 2011) and provides efficient incorporation of isotopically labeled amino acids, with similar yields per L versus unlabeled expression in rich media. Gossert et al. identified the presence of unlabeled amino acids in the yeastolate of the growth medium as a major limitation in isotope labeling using BV-infected insect cells. By reducing the amount of yeastolate in the growth medium ten-fold, a significant improvement in labeling efficiency was demonstrated, while maintaining good protein expression yield. We report an alternate approach to improve isotope labeling efficiency using BV-infected insect cells namely by replacing the yeast extracts in the medium with dialyzed yeast extracts to reduce the amount of low molecular weight peptides and amino acids. We report the residual levels of amino acids in various media formulations and the amino acid consumption during fermentation, as determined by NMR. While direct replacement of yeastolate with dialyzed yeastolate delivered moderately lower isotope labeling efficiencies compared to the use of ten-fold diluted undialized yeastolate, we show that the use of dialyzed yeastolate combined with a ten-fold dilution delivered enhanced isotope labeling efficiency and at least a comparable level of protein expression yield, all at a scale which economizes use of these costly reagents.


Asunto(s)
Marcaje Isotópico/métodos , Aminoácidos/análisis , Aminoácidos/química , Animales , Baculoviridae , Antígenos CD4/biosíntesis , Antígenos CD4/química , Antígenos CD4/aislamiento & purificación , Isótopos de Carbono , Medios de Cultivo/análisis , Medios de Cultivo/química , Quinasa 1 de Adhesión Focal/biosíntesis , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/aislamiento & purificación , Isótopos de Nitrógeno , Resonancia Magnética Nuclear Biomolecular , Biosíntesis de Proteínas , Células Sf9 , Spodoptera
15.
Proc Natl Acad Sci U S A ; 114(15): 3933-3938, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28348210

RESUMEN

Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase involved in development and human disease, including cancer. It is currently thought that the four-point one, ezrin, radixin, moesin (FERM)-kinase domain linker, which contains autophosphorylation site tyrosine (Y) 397, is not required for in vivo FAK function until late midgestation. Here, we directly tested this hypothesis by generating mice with FAK Y397-to-phenylalanine (F) mutations in the germline. We found that Y397F embryos exhibited reduced mesodermal fibronectin (FN) and osteopontin expression and died during mesoderm development akin to FAK kinase-dead mice. We identified myosin-1E (MYO1E), an actin-dependent molecular motor, to interact directly with the FAK FERM-kinase linker and induce FAK kinase activity and Y397 phosphorylation. Active FAK in turn accumulated in the nucleus where it led to the expression of osteopontin and other FN-type matrix in both mouse embryonic fibroblasts and human melanoma. Our data support a model in which FAK Y397 autophosphorylation is required for FAK function in vivo and is positively regulated by MYO1E.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Melanoma/metabolismo , Miosinas/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Pérdida del Embrión/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/genética , Humanos , Melanoma/patología , Mesodermo/embriología , Ratones Mutantes , Miosina Tipo I , Miosinas/química , Miosinas/genética , Osteopontina/genética , Osteopontina/metabolismo , Fosforilación , Embarazo , Dominios Proteicos , Neoplasias Cutáneas/patología , Tirosina/metabolismo
16.
J Phys Chem B ; 121(15): 3523-3535, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28124908

RESUMEN

Focal adhesion kinase (FAK) is a multidomain protein (FERM-kinase-FAT) with important signaling functions in the regulation of cell-substrate adhesions. Its inactive, autoinhibited form is recruited from the cytoplasm to the plasma membrane, where it becomes activated at PIP2 enriched regions. To elucidate the molecular basis of activation, we performed a systematic screening of binding orientations of FAK's autoinhibited FERM-kinase complex, as well as of the dissociated FERM and kinase domains alone, on model plasma membranes using coarse-grained scFix MARTINI simulations, partially corroborated by atomistic MD simulations. The proteins adopted many more different orientations than previously thought. The presence of PIP2 tuned and narrowed the complex map of competing interfacial orientations. The dissociated FERM domain most frequently interacted with the membrane through its autoinhibitory interface rather than with the "basic patch" residues. These findings suggest a PIP2-dependent activation mechanism in which membrane binding of the dissociated FERM domain competes with the rebinding of the kinase domain. This competition could promote FAK autophosphorylation on Y397 and subsequent Src binding. The orientation of peripheral proteins at membranes is crucial to understand cell adhesion processes and is furthermore required to exploit steered molecular dynamics to predict how tensile forces might switch their active states.


Asunto(s)
Quinasa 1 de Adhesión Focal/química , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Sitios de Unión , Humanos , Conformación Proteica
17.
Biochim Biophys Acta Proteins Proteom ; 1865(1): 43-54, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27718363

RESUMEN

Therapeutic protein kinase inhibitors are designed on the basis of kinase structures. Here, we define intrinsically disordered regions (IDRs) in structurally hybrid kinases. We reveal that 65% of kinases have an IDR adjacent to their kinase domain (KD). These IDRs are evolutionarily more conserved than IDRs distant to KDs. Strikingly, 36 kinases have adjacent IDRs extending into their KDs, defining a unique structural and functional subset of the kinome. Functional network analysis of this subset of the kinome uncovered FAK1 as topologically the most connected hub kinase. We identify that KD-flanking IDR of FAK1 is more conserved and undergoes more post-translational modifications than other IDRs. It preferentially interacts with proteins regulating scaffolding and kinase activity, which contribute to cytoskeletal remodeling. In summary, spatially and evolutionarily conserved IDRs in kinases may influence their functions, which can be exploited for targeted therapies in diseases including those that involve aberrant cytoskeletal remodeling.


Asunto(s)
Citoesqueleto/metabolismo , Quinasa 1 de Adhesión Focal/química , Citoesqueleto/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Conformación Proteica , Procesamiento Proteico-Postraduccional
18.
Cancer Lett ; 385: 1-11, 2017 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-27816491

RESUMEN

The natural bioactive compound damnacanthal inhibits several tyrosine kinases. Herein, we show that -in fact- damancanthal is a multi kinase inhibitor. A docking and molecular dynamics simulation approach allows getting further insight on the inhibitory effect of damnacanthal on three different kinases: vascular endothelial growth factor receptor-2, c-Met and focal adhesion kinase. Several of the kinases targeted and inhibited by damnacanthal are involved in angiogenesis. Ex vivo and in vivo experiments clearly demonstrate that, indeed, damnacanthal is a very potent inhibitor of angiogenesis. A number of in vitro assays contribute to determine the specific effects of damnacanthal on each of the steps of the angiogenic process, including inhibition of tubulogenesis, endothelial cell proliferation, survival, migration and production of extracellular matrix remodeling enzyme. Taken altogether, these results suggest that damancanthal could have potential interest for the treatment of cancer and other angiogenesis-dependent diseases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antraquinonas/farmacología , Membrana Corioalantoides/irrigación sanguínea , Receptores ErbB/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Inhibidores de la Angiogénesis/química , Animales , Antraquinonas/química , Bovinos , Adhesión Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Receptores ErbB/química , Receptores ErbB/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Neovascularización Fisiológica/efectos de los fármacos , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-met/química , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Pez Cebra/embriología
19.
Structure ; 24(8): 1301-1310, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27427476

RESUMEN

Focal adhesion kinase (FAK) has emerged as a mediator of mechanotransduction in cardiomyocytes, regulating gene expression during hypertrophic remodeling. However, how FAK signaling is relayed onward to the nucleus is unclear. Here, we show that FAK interacts with and regulates myocyte enhancer factor 2 (MEF2), a master cardiac transcriptional regulator. In cardiomyocytes exposed to biomechanical stimulation, FAK accumulates in the nucleus, binds to and upregulates the transcriptional activity of MEF2 through an interaction with the FAK focal adhesion targeting (FAT) domain. In the crystal structure (2.9 Å resolution), FAT binds to a stably folded groove in the MEF2 dimer, known to interact with regulatory cofactors. FAK cooperates with MEF2 to enhance the expression of Jun in cardiomyocytes, an important component of hypertrophic response to mechanical stress. These findings underscore a connection between the mechanotransduction involving FAK and transcriptional regulation by MEF2, with potential relevance to the pathogenesis of cardiac disease.


Asunto(s)
Quinasa 1 de Adhesión Focal/química , Mecanotransducción Celular , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-jun/química , Transcripción Genética , Secuencias de Aminoácidos , Animales , Animales Recién Nacidos , Sitios de Unión , Línea Celular , Núcleo Celular/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Cinética , Factores de Transcripción MEF2/química , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Modelos Moleculares , Miocitos Cardíacos/citología , Cultivo Primario de Células , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
Biochim Biophys Acta ; 1860(7): 1388-94, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27033120

RESUMEN

The regulation of focal adhesion kinase (FAK) involves phosphorylation and multiple interactions with other signaling proteins. Some of these pathways are relevant for nervous system functions such as branching, axonal guidance, and plasticity. In this study, we screened mouse brain to identify FAK-interactive proteins and phosphorylatable residues as a first step to address the neuronal functions of this kinase. Using mass spectrometry analysis, we identified new phosphorylated sites (Thr 952, Thr 1048, and Ser 1049), which lie in the FAT domain; and putative new partners for FAK, which include cytoskeletal proteins such as drebrin and MAP 6, adhesion regulators such as neurabin-2 and plakophilin 1, and synapse-associated proteins such as SynGAP and a NMDA receptor subunit. Our findings support the participation of brain-localized FAK in neuronal plasticity.


Asunto(s)
Encéfalo/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Convulsiones/enzimología , Espectrometría de Masas en Tándem , Animales , Animales Recién Nacidos , Sitios de Unión , Encéfalo/fisiopatología , Dominio Catalítico , Cromatografía Liquida , Modelos Animales de Enfermedad , Activación Enzimática , Quinasa 1 de Adhesión Focal/química , Inmunoprecipitación , Ratones , Plasticidad Neuronal , Pentilenotetrazol , Fosforilación , Unión Proteica , Convulsiones/fisiopatología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...