Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Theranostics ; 11(16): 8092-8111, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335982

RESUMEN

Active c-Src non-receptor tyrosine kinase localizes to the plasma membrane via N-terminal lipid modification. Membranous c-Src causes cancer initiation and progression. Even though transmembrane 4 L six family member 5 (TM4SF5), a tetraspan(in), can be involved in this mechanism, the molecular and structural influence of TM4SF5 on c-Src remains unknown. Methods: Here, we investigated molecular and structural details by which TM4SF5 regulated c-Src devoid of its N-terminus and how cell-penetrating peptides were able to interrupt c-Src activation via interference of c-Src-TM4SF5 interaction in hepatocellular carcinoma models. Results: The TM4SF5 C-terminus efficiently bound the c-Src SH1 kinase domain, efficiently to the inactively-closed form. The complex involved protein tyrosine phosphatase 1B able to dephosphorylate Tyr530. The c-Src SH1 domain alone, even in a closed form, bound TM4SF5 to cause c-Src Tyr419 and FAK Y861 phosphorylation. Homology modeling and molecular dynamics simulation studies predicted the directly interfacing residues, which were further validated by mutational studies. Cell penetration of TM4SF5 C-terminal peptides blocked the interaction of TM4SF5 with c-Src and prevented c-Src-dependent tumor initiation and progression in vivo. Conclusions: Collectively, these data demonstrate that binding of the TM4SF5 C-terminus to the kinase domain of inactive c-Src leads to its activation. Because this binding can be abolished by cell-penetrating peptides containing the TM4SF5 C-terminus, targeting this direct interaction may be an effective strategy for developing therapeutics that block the development and progression of hepatocellular carcinoma.


Asunto(s)
Proteína Tirosina Quinasa CSK/metabolismo , Carcinoma Hepatocelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Tirosina Quinasa CSK/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Genes src/genética , Genes src/fisiología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Péptidos/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Tetraspaninas/genética , Tetraspaninas/metabolismo
2.
Ann Med ; 53(1): 596-610, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33830879

RESUMEN

PURPOSE: This study aims to identify potential prognostic biomarkers of bladder cancer (BCa) based on large-scale multi-omics data and investigate the role of SRC in improving predictive outcomes for BCa patients and those receiving immune checkpoint therapies (ICTs). METHODS: Large-scale multi-comic data were enrolled from the Cancer Proteome Atlas, the Cancer Genome Atlas and gene expression omnibus based on machining-learning methods. Immune infiltration, survival and other statistical analyses were implemented using R software in cancers (n = 12,452). The predictive value of SRC was performed in 81 BCa patients receiving ICT from aa validation cohort (n = 81). RESULTS: Landscape of novel candidate prognostic protein signatures of BCa patients was identified. Differential BECLIN, EGFR, PKCALPHA, ANNEXIN1, AXL and SRC expression significantly correlated with the outcomes for BCa patients from multiply cohorts (n = 906). Notably, risk score of the integrated prognosis-related proteins (IPRPs) model exhibited high diagnostic accuracy and consistent predictive ability (AUC = 0.714). Besides, we tested the clinical relevance of baseline SRC protein and mRNA expression in two independent confirmatory cohorts (n = 566) and the prognostic value in pan-cancers. Then, we found that elevated SRC expression contributed to immunosuppressive microenvironment mediated by immune checkpoint molecules of BCa and other cancers. Next, we validated SRC expression as a potential biomarker in predicting response to ICT in 81 BCa patient from FUSCC cohort, and found that expression of SRC in the baseline tumour tissues correlated with improved survival benefits, but predicts worse ICT response. CONCLUSION: This study first performed the large-scale multi-omics analysis, distinguished the IPRPs (BECLIN, EGFR, PKCALPHA, SRC, ANNEXIN1 and AXL) and revealed novel prediction model, outperforming the currently traditional prognostic indicators for anticipating BCa progression and better clinical strategies. Additionally, this study provided insight into the importance of biomarker SRC for better prognosis, which may inversely improve predictive outcomes for patients receiving ICT and enable patient selection for future clinical treatment.


Asunto(s)
Inmunidad Adaptativa/genética , Genes src/genética , Genómica/estadística & datos numéricos , Inmunoterapia , Neoplasias de la Vejiga Urinaria/genética , Anexina A1/metabolismo , Área Bajo la Curva , Beclina-1/metabolismo , Biomarcadores de Tumor/genética , Bases de Datos Genéticas , Receptores ErbB/metabolismo , Expresión Génica/genética , Genómica/métodos , Humanos , Aprendizaje Automático , Selección de Paciente , Valor Predictivo de las Pruebas , Pronóstico , Modelos de Riesgos Proporcionales , Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factores de Riesgo , Análisis de Supervivencia , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Tirosina Quinasa del Receptor Axl
3.
J Neurosci ; 41(17): 3808-3821, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33727334

RESUMEN

To assemble the functional circuits of the nervous system, the neuronal axonal growth cones must be precisely guided to their proper targets, which can be achieved through cell-surface guidance receptor activation by ligand binding in the periphery. We investigated the function of paxillin, a focal adhesion protein, as an essential growth cone guidance intermediary in the context of spinal lateral motor column (LMC) motor axon trajectory selection in the limb mesenchyme. Using in situ mRNA detection, we first show paxillin expression in LMC neurons of chick and mouse embryos at the time of spinal motor axon extension into the limb. Paxillin loss-of-function and gain-of-function using in ovo electroporation in chick LMC neurons, of either sex, perturbed LMC axon trajectory selection, demonstrating an essential role of paxillin in motor axon guidance. In addition, a neuron-specific paxillin deletion in mice led to LMC axon trajectory selection errors. We also show that knocking down paxillin attenuates the growth preference of LMC neurites against ephrins in vitro, and erythropoietin-producing human hepatocellular (Eph)-mediated retargeting of LMC axons in vivo, suggesting paxillin involvement in Eph-mediated LMC motor axon guidance. Finally, both paxillin knockdown and ectopic expression of a nonphosphorylable paxillin mutant attenuated the retargeting of LMC axons caused by Src overexpression, implicating paxillin as a Src target in Eph signal relay in this context. In summary, our findings demonstrate that paxillin is required for motor axon guidance and suggest its essential role in the ephrin-Eph signaling pathway resulting in motor axon trajectory selection.SIGNIFICANCE STATEMENT During the development of neural circuits, precise connections need to be established among neurons or between neurons and their muscle targets. A protein family found in neurons, Eph, is essential at different stages of neural circuit formation, including nerve outgrowth and pathfinding, and is proposed to mediate the onset and progression of several neurodegenerative diseases, such as Alzheimer's disease. To investigate how Ephs relay their signals to mediate nerve growth, we investigated the function of a molecule called paxillin and found it important for the development of spinal nerve growth toward their muscle targets, suggesting its role as an effector of Eph signals. Our work could thus provide new information on how neuromuscular connectivity is properly established during embryonic development.


Asunto(s)
Axones/fisiología , Paxillin/fisiología , Médula Espinal/crecimiento & desarrollo , Animales , Orientación del Axón/fisiología , Embrión de Pollo , Electroporación , Efrinas/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Genes src/genética , Humanos , Masculino , Ratones , MicroARNs/genética , Neuronas Motoras/fisiología , Mutación/genética , Neuritas/fisiología , Médula Espinal/citología
4.
Sci Rep ; 9(1): 4819, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894630

RESUMEN

Lung cancer is the foremost cause of cancer related deaths in the U.S. It is a heterogeneous disease composed of genetically and phenotypically distinct tumor cells surrounded by heterotypic cells and extracellular matrix dynamically interacting with the tumor cells. Research in lung cancer is often restricted to patient-derived tumor specimens, in vitro cell cultures and limited animal models, which fail to capture the cellular or microenvironment heterogeneity of the tumor. Therefore, our knowledge is primarily focused on cancer-cell autonomous aberrations. For a fundamental understanding of lung cancer progression and an exploration of therapeutic options, we focused our efforts to develop an Ex Vivo Tumor platform to culture tumors in 3D matrices, which retains tumor cell heterogeneity arising due to in vivo selection pressure and environmental influences and recapitulate responses of tumor cells to external manipulations. To establish this model, implanted syngeneic murine tumors from a mutant KRAS/p53 model were harvested to yield multicellular tumor aggregates followed by culture in 3D extracellular matrices. Using this system, we identified Src signaling as an important driver of invasion and metastasis in lung cancer and demonstrate that EVTs are a robust experimental tool bridging the gap between conventional in vitro and in vivo models.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Transición Epitelial-Mesenquimal/genética , Genes src/genética , Neoplasias Pulmonares/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Matriz Extracelular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pulmonares/patología , Ratones , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Ratas , Transducción de Señal/genética , Esferoides Celulares/patología , Microambiente Tumoral/genética
5.
Int J Mol Sci ; 20(5)2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30845661

RESUMEN

Glioblastoma (GBM) is the most aggressive type of brain tumor, with an overall survival of 17 months under the current standard of care therapy. CD99, an over-expressed transmembrane protein in several malignancies, has been considered a potential target for immunotherapy. To further understand this potentiality, we analyzed the differential expression of its two isoforms in human astrocytoma specimens, and the CD99 involved signaling pathways in glioma model U87MG cell line. CD99 was also analyzed in GBM molecular subtypes. Whole transcriptomes by RNA-Seq of CD99-siRNA, and functional in vitro assays in CD99-shRNA, that are found in U87MG cells, were performed. Astrocytoma of different malignant grades and U87MG cells only expressed CD99 isoform 1, which was higher in mesenchymal and classical than in proneural GBM subtypes. Genes related to actin dynamics, predominantly to focal adhesion, and lamellipodia/filopodia formation were down-regulated in the transcriptome analysis, when CD99 was silenced. A decrease in tumor cell migration/invasion, and dysfunction of focal adhesion, were observed in functional assays. In addition, a striking morphological change was detected in CD99-silenced U87MG cells, further corroborating CD99 involvement in actin cytoskeleton rearrangement. Inhibiting the overexpressed CD99 may improve resectability and decrease the recurrence rate of GBM by decreasing tumor cells migration and invasion.


Asunto(s)
Antígeno 12E7/genética , Antígeno 12E7/metabolismo , Neoplasias Encefálicas/genética , Perfilación de la Expresión Génica/métodos , Glioblastoma/genética , Regulación hacia Arriba , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes src/genética , Glioblastoma/metabolismo , Humanos , Invasividad Neoplásica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/farmacología , Análisis de Secuencia de ARN
6.
Oncogene ; 38(21): 4015-4027, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30692633

RESUMEN

Aside from its well-known nuclear routes of signaling, estrogen also mediates its effects through cytoplasmic signaling. Estrogen signaling involves numerous posttranslational modifications of its receptor ERα, the best known being phosphorylation. Our research group previously showed that upon estrogen stimulation, ERα is methylated on residue R260 and forms the mERα/Src/PI3K complex, central to the rapid transduction of nongenomic estrogen signals. Regulation of ERα signaling via its phosphorylation by growth factors is well recognized, and we wondered whether they could also trigger ERα methylation (mERα). Here, we found that IGF-1 treatment of MCF-7 cells induced rapid ERα methylation by the arginine methyltransferase PRMT1 and triggered the binding of mERα to IGF-1R. Mechanistically, we showed that PRMT1 bound constitutively to IGF-1R and that PRMT1 became activated upon IGF-1 stimulation. Moreover, we found that expression or pharmacological inhibition of PRMT1 impaired mERα and IGF-1 signaling. Our findings were substantiated in a cohort of breast tumors in which IGF-1R expression was positively correlated with ERα/Src and ERα/PI3K expression, hallmarks of nongenomic estrogen signaling, reinforcing the link between IGF-1R and mERα. Altogether, these results provide a new insight into ERα and IGF-1R interference, and open novel perspectives for combining endocrine therapies with PRMT1 inhibitors in ERα-positive tumors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Femenino , Genes src/genética , Humanos , Células MCF-7 , Metilación , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica/fisiología , Receptor IGF Tipo 1/metabolismo
7.
Cell Death Dis ; 9(9): 877, 2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158525

RESUMEN

Many stress conditions including chemotherapy treatment is known to activate Src and under certain condition Src can induce the apoptotic signal via c-Jun N-terminal kinase (JNK) activation. Here we report that the newly synthesized ß-phenylacrylic acid derivatives, MHY791 and MHY1036 (MHYs), bind to epidermal growth factor receptor (EGFR) tyrosine kinase domains and function as EGFR inhibitors, having anti-cancer activities selectively in wild-type KRAS colon cancer. Mechanistically, MHYs-induced Src/JNK activation which enhanced their pro-apoptotic effects and therefore inhibition of Src by the chemical inhibitor PP2 or Src siRNA abolished the response. In addition, MHYs generated reactive oxygen species and increased ER stress, and pretreatment with antioxidant-inhibited MHY-induced ER stress, Src activation, and apoptosis. Furthermore, the irreversible EGFR inhibitor PD168393 also activated Src while the reversible EGFR inhibitor gefitinib showed the opposite effect, indicating that MHYs are the irreversible EGFR inhibitor. Collectively, Src can play a key role in apoptosis induced by the novel EGFR inhibitor MHYs, suggesting that activation of Src might prove effective in treating EGFR/wild-type KRAS colon cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Genes src/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Familia-src Quinasas/genética , Apoptosis/genética , Células CACO-2 , Línea Celular Tumoral , Receptores ErbB/genética , Gefitinib/farmacología , Células HCT116 , Células HT29 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/genética , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Cancer Res ; 78(11): 2825-2838, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29531159

RESUMEN

Posttranslational modifications of mammalian c-Src N-terminal and C-terminal domains regulate distinct functions. Myristoylation of G2 controls its cell membrane association and phosphorylation of Y419/Y527 controls its activation or inactivation, respectively. We provide evidence that Src-cell membrane association-dissociation and catalytic activation-inactivation are both regulated by acetylation. In EGF-treated cells, CREB binding protein (CBP) acetylates an N-terminal lysine cluster (K5, K7, and K9) of c-Src to promote dissociation from the cell membrane. CBP also acetylates the C-terminal K401, K423, and K427 of c-Src to activate intrinsic kinase activity for STAT3 recruitment and activation. N-terminal domain phosphorylation (Y14, Y45, and Y68) of STAT3 by c-Src activates transcriptionally active dimers of STAT3. Moreover, acetyl-Src translocates into nuclei, where it forms the Src-STAT3 enhanceosome for gene regulation and cancer cell proliferation. Thus, c-Src acetylation in the N-terminal and C-terminal domains play distinct roles in Src activity and regulation.Significance: CBP-mediated acetylation of lysine clusters in both the N-terminal and C-terminal regions of c-Src provides additional levels of control over STAT3 transcriptional activity. Cancer Res; 78(11); 2825-38. ©2018 AACR.


Asunto(s)
Carcinogénesis/genética , Genes src/genética , Procesamiento Proteico-Postraduccional/genética , Factor de Transcripción STAT3/genética , Acetilación , Animales , Proteína de Unión a CREB/genética , Proteína Tirosina Quinasa CSK , Línea Celular , Línea Celular Tumoral , Núcleo Celular/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Células MCF-7 , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Fosforilación/genética , Proteínas Tirosina Quinasas/genética , Transactivadores/genética , Transcripción Genética/genética , Familia-src Quinasas/genética
9.
Neoplasia ; 20(3): 233-243, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29444487

RESUMEN

Cross talk of stromal-epithelial cells plays an essential role in both normal development and tumor initiation and progression. Fibroblast growth factor (FGF)-FGF receptor (FGFR)-Src kinase axis is one of the major signal transduction pathways to mediate this cross talk. Numerous genomic studies have demonstrated that expression levels of FGFR/Src are deregulated in a variety of cancers including prostate cancer; however, the role that paracrine FGF (from stromal cells) plays in dysregulated expression of epithelial FGFRs/Src and tumor progression in vivo is not well evaluated. In this study, we demonstrate that ectopic expression of wild-type FGFR1/2 or Src kinase in epithelial cells was not sufficient to initiate prostate tumorigenesis under a normal stromal microenvironment in vivo. However, paracrine FGF10 synergized with ectopic expression of epithelial FGFR1 or FGFR2 to induce epithelial-mesenchymal transition. Additionally, paracrine FGF10 sensitized FGFR2-transformed epithelial cells to initiate prostate tumorigenesis. Next, paracrine FGF10 also synergized with overexpression of epithelial Src kinase to high-grade tumors. But loss of the myristoylation site in Src kinase inhibited paracrine FGF10-induced prostate tumorigenesis. Loss of myristoylation alters Src levels in the cell membrane and inhibited FGF-mediated signaling including inhibition of the phosphotyrosine pattern and FAK phosphorylation. Our study demonstrates the potential tumor progression by simultaneous deregulation of proteins in the FGF/FGFRs/Src signal axis and provides a therapeutic strategy of targeting myristoylation of Src kinase to interfere with the tumorigenic process.


Asunto(s)
Transformación Celular Neoplásica/patología , Factores de Crecimiento de Fibroblastos/genética , Genes src/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Oncogenes/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/genética , Células del Estroma/metabolismo , Células del Estroma/patología
10.
J BUON ; 22(5): 1097-1106, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29135089

RESUMEN

PURPOSE: The FAK/Src/Paxillin (PXN) axis has been implicated in malignant transformation, tumor growth, progression and metastasis. The present study aimed to assess FAK/Src/PXN protein expression in both primary and liver metastatic sites of colorectal adenocarcinoma (CRC). METHODS: FAK, Src and p-PXN expression was assessed immunohistochemically on 32 primary CRCs and their corresponding liver metastases, being also analyzed in relation with clinicopathological characteristics and patient survival. RESULTS: FAK, Src and p-PXN expression was significantly decreased in liver metastasis compared to matched paired primary CRCs (p<0.01). Increased FAK expression in primary CRCs was significantly associated with poor histological grade and advanced disease stage (p=0.0330 and p=0.0204, respectively). Increased Src expression in primary colorectal tumors was significantly associated with the presence of lymph node metastasis (p=0.0325), while elevated p-PXN expression with poor histological grade (p=0.0284). CONCLUSIONS: FAK, Src and p-PXN appear to play a role in the pathophysiological aspects of CRC. The lower expression of these proteins in liver metastasis compared to the primary CRC could significantly impact the choice of a novel therapeutic agent according to the disease stage.


Asunto(s)
Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/genética , Quinasa 1 de Adhesión Focal/metabolismo , Genes src/genética , Neoplasias Hepáticas/secundario , Paxillin/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Humanos , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad
11.
J Pharmacol Exp Ther ; 363(3): 428-443, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29021381

RESUMEN

Despite encouraging clinical results with sorafenib monotherapy in patients with KRAS-mutant non-small-cell lung cancer (NSCLC), the overall survival benefit of this drug is limited by the inevitable development of acquired resistance. The exact mechanism underlying acquired sorafenib resistance in KRAS-mutant NSCLC is unclear. In this study, the mechanism of acquired sorafenib resistance was explored using a biologically relevant xenograft model, which was established by using the A549 human lung adenocarcinoma cell line and an in vivo-derived, sorafenib-resistant A549 subline (A549/SRFres). Results from the initial study demonstrated that sorafenib treatment significantly decreased E-cadherin (P < 0.05) levels but significantly increased matrix metallopeptidase 9 (MMP9) levels (P < 0.01) in A549/SRFres tumors, whereas expression levels of phospho-protein kinase B (AKT), phospho-focal adhesion kinase (FAK), and phospho-Src were elevated in sorafenib-treated A549 and A549/SRFres tumors. We next examined whether concomitant dasatinib treatment could overcome acquired sorafenib resistance by blocking the FAK/Src escape route that mediates resistance. Despite the observed in vitro synergy between sorafenib and dasatinib, the in vivo antitumor effect of half-dose sorafenib-dasatinib combination therapy was inferior to that of the full-dose sorafenib treatment. Although the sorafenib-dasatinib combination effectively inhibited Src and AKT phosphorylation, it did not block the Y576/577-FAK phosphorylation, nor did it decrease vimentin protein expression; unexpectedly, it increased Y397-FAK phosphorylation and MMP9 protein expression in tumors. These results suggest that acquired sorafenib resistance in KRAS-mutant A549 xenografts involves the compensatory activation of FAK and Src, and Src inhibition alone is insufficient to diminish sorafenib-promoted epithelial-mesenchymal transition process and invasive potentials in tumors.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Proteína-Tirosina Quinasas de Adhesión Focal/biosíntesis , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Genes src/genética , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Células A549 , Animales , Dasatinib/farmacología , Sinergismo Farmacológico , Activación Enzimática , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Desnudos , Niacinamida/farmacología , Proteína Oncogénica v-akt/biosíntesis , Proteína Oncogénica v-akt/genética , Sorafenib , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mediators Inflamm ; 2017: 8123281, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28804222

RESUMEN

Epithelial-mesenchymal transition (EMT) is a biological process that allows epithelial cells to assume a mesenchymal cell phenotype. EMT is considered as a therapeutic target for several persistent inflammatory airway diseases related to tissue remodeling. Herein, we investigated the role of endoplasmic reticulum (ER) stress and c-Src in TGF-ß1-induced EMT. A549 cells, primary nasal epithelial cells (PNECs), and inferior nasal turbinate organ cultures were exposed to 4-phenylbutylic acid (4PBA) or PP2 and then stimulated with TGF-ß1. We found that E-cadherin, vimentin, fibronectin, and α-SMA expression was increased in nasal polyps compared to inferior turbinates. TGF-ß1 increased the expression of EMT markers such as E-cadherin, fibronectin, vimentin, and α-SMA and ER stress markers (XBP-1s and GRP78), an effect that was blocked by PBA or PP2 treatment. 4-PBA and PP2 also blocked the effect of TGF-ß1 on migration of A549 cells and suppressed TGF-ß1-induced expression of EMT markers in PNECs and organ cultures of inferior turbinate. In conclusion, we demonstrated that 4PBA inhibits TGF-ß1-induced EMT via the c-Src pathway in A549 cells, PNECs, and inferior turbinate organ cultures. These results suggest an important role for ER stress and a diverse role for TGF-ß1 in upper airway chronic inflammatory disease such as CRS.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Genes src/fisiología , Factor de Crecimiento Transformador beta1/farmacología , Células A549 , Movimiento Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Genes src/genética , Humanos , Pólipos Nasales/metabolismo , Técnicas de Cultivo de Órganos , Transducción de Señal/efectos de los fármacos
13.
J Endocrinol ; 233(2): 175-186, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28223314

RESUMEN

Transgenic mice overexpressing growth hormone (GH) show increased hepatic protein content of the epidermal growth factor receptor (EGFR), which is broadly associated with cell proliferation and oncogenesis. However, chronically elevated levels of GH result in desensitization of STAT-mediated EGF signal and similar response of ERK1/2 and AKT signaling to EGF compared to normal mice. To ascertain the mechanisms involved in GH attenuation of EGF signaling and the consequences on cell cycle promotion, phosphorylation of signaling mediators was studied at different time points after EGF stimulation, and induction of proteins involved in cell cycle progression was assessed in normal and GH-overexpressing transgenic mice. Results from kinetic studies confirmed the absence of STAT3 and 5 activation and comparable levels of ERK1/2 phosphorylation upon EGF stimulation, which was associated with diminished or similar induction of c-MYC, c-FOS, c-JUN, CYCLIN D1 and CYCLIN E in transgenic compared to normal mice. Accordingly, kinetics of EGF-induced c-SRC and EGFR phosphorylation at activating residues demonstrated that activation of these proteins was lower in the transgenic mice with respect to normal animals. In turn, EGFR phosphorylation at serine 1046/1047, which is implicated in the negative regulation of the receptor, was increased in the liver of GH-overexpressing transgenic mice both in basal conditions and upon EGF stimulus. Increased basal phosphorylation and activation of the p38-mitogen-activated protein kinase might account for increased Ser 1046/1047 EGFR. Hyperphosphorylation of EGFR at serine residues would represent a compensatory mechanism triggered by chronically elevated levels of GH to mitigate the proliferative response induced by EGF.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Regulación de la Expresión Génica/fisiología , Hormona del Crecimiento/metabolismo , Transducción de Señal/fisiología , Animales , Receptores ErbB/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genes src/genética , Genes src/fisiología , Hormona del Crecimiento/genética , Humanos , Hígado/metabolismo , Ratones , Ratones Transgénicos , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Mol Biol Cell ; 27(24): 3926-3936, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27733622

RESUMEN

The interactions of Src family kinases (SFKs) with the plasma membrane are crucial for their activity. They depend on their fatty-acylated N-termini, containing N-myristate and either a polybasic cluster (in Src) or palmitoylation sites (e.g., Fyn). To investigate the roles of these moieties in SFK membrane association, we used fluorescence recovery after photobleaching beam-size analysis to study the membrane interactions of c-Src-GFP (green fluorescent protein) or Fyn-GFP fatty-acylation mutants. Our studies showed for the first time that the membrane association of Fyn is more stable than that of Src, an effect lost in a Fyn mutant lacking the palmitoylation sites. Unexpectedly, Src-S3C/S6C (containing cysteines at positions 3/6, which are palmitoylated in Fyn) exhibited fast cytoplasmic diffusion insensitive to palmitoylation inhibitors, suggesting defective fatty acylation. Further replacement of the charged Lys-5 by neutral Gln to resemble Fyn (Src-S3C/S6C/K5Q) restored Fyn-like membrane interactions, indicating that Lys-5 in the context of Src-S3C/S6C interferes with its myristoylation/palmitoylation. This was validated by direct myristoylation and palmitoylation studies, which indicated that the residue at position 5 regulates the membrane interactions of Src versus Fyn. Moreover, the palmitoylation levels correlated with targeting to detergent-resistant membranes (rafts) and to caveolin-1. Palmitoylation-dependent preferential containment of Fyn in rafts may contribute to its lower transformation potential.


Asunto(s)
Genes src/genética , Genes src/fisiología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Acilación , Secuencia de Aminoácidos , Animales , Células COS , Proteína Tirosina Quinasa CSK , Caveolina 1/metabolismo , Membrana Celular/metabolismo , Chlorocebus aethiops , Cisteína/metabolismo , Proteínas Fluorescentes Verdes , Lipoilación , Proteínas de la Membrana , Membranas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fyn/genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
15.
Avian Pathol ; 45(2): 202-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27100152

RESUMEN

To understand the cytogenetic characteristics of acute fibrosarcoma in chickens infected with the subgroup J avian leukosis virus associated with the v-src oncogene, we performed a karyotype analysis of fibrosarcoma cell cultures. Twenty-nine of 50 qualified cell culture spreads demonstrated polyploidy of some macrochromosomes, 21 of which were trisomic for chromosome 7, and others were trisomic for chromosomes 3, 4, 5 (sex chromosome w), and 10. In addition, one of them was trisomic for both chromosome 7 and the sex chromosome 5 (w). In contrast, no aneuploidy was found for 10 macrochromosomes of 12 spreads of normal chicken embryo fibroblast cells, although aneuploidy for some microchromosomes was demonstrated in five of the 12 spreads. The cytogenetic mosaicism or polymorphism of the aneuploidy in the acute fibrosarcoma described in this study suggests that the analysed cells are polyclonal.


Asunto(s)
Virus de la Leucosis Aviar/genética , Leucosis Aviar/virología , Pollos/virología , Aberraciones Cromosómicas , Fibrosarcoma/veterinaria , Genes src/genética , Enfermedad Aguda , Animales , Leucosis Aviar/genética , Virus de la Leucosis Aviar/aislamiento & purificación , Embrión de Pollo , Femenino , Fibrosarcoma/genética , Fibrosarcoma/virología , Cariotipo , Cariotipificación/veterinaria , Polimorfismo Genético
16.
Anesth Analg ; 122(3): 871-881, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26720617

RESUMEN

BACKGROUND: The elusiveness of pain mechanisms is a major impediment in developing effective clinical treatments. We examined whether the signal regulatory protein α1 (SIRPα1)-activated spinal Src homology-2 domain-containing protein tyrosine phosphatase 2 (SHP2)/Src cascade and the downstream GluN2B phosphorylation play a role in inflammatory pain. METHODS: At hour 3 and days 1, 3, 5, and 10 after the intraplantar injection of complete Freund adjuvant (CFA), we assessed paw withdrawal latency using the Hargreaves test and analyzed dorsal horn samples (L4-L5) by Western blotting and immunoprecipitation. RESULTS: Intraplantar CFA injection provoked the behavioral hyperalgesia in the ipsilateral hind-paw along with SIRPα1, phosphorylated SHP2 (pSHP2), phosphorylated Src (pSrc), and phosphorylated GluN2B expressions and total SHP2 (tSHP2)-SIRPα1/pSHP2/pSrc and total Src (tSrc)-SIRPα1/pSHP2/pSrc coprecipitation in the ipsilateral dorsal horn. Although both of them failed to show an effect on CFA-enhanced SIRPα1 expression, spinal administration with SIRPα1-neutralizing antibody (10, 50, and 100 µg, 10 µL) and 8-Hydroxy-7-[(6-sulfo-2-naphthyl)azo]-5-quinolinesulfonic acid (NSC 8787; an SHP2 antagonist, 1, 10, and 100 µM, 10 µL) dose-dependently attenuated the behavioral hyperalgesia, SHP2 and Src phosphorylation, and tSHP2-SIRPα1/pSHP2/pSrc coprecipitation at day 1 after CFA injection. Intrathecal application of 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2; a Src-family kinase inhibitor, 10, 30, and 50 nM, 10 µL) exhibited a similar effect as these agents, except that it failed to ameliorate CFA-enhanced SHP2 phosphorylation and tSHP2-SIRPα1/pSHP2 coprecipitation. CONCLUSIONS: CFA-induced spinal SIRPα1 expression, which triggers SHP2, and Src phosphorylation, which subsequently induced pSrc-GluN2B interaction to mediate the GluN2B activation, contribute to spinal plasticity underlying the maintenance of inflammatory pain. These findings provide a possible strategy for pain relief by targeting to spinal SIRPα1-SHP2 coupling.


Asunto(s)
Genes src/genética , Inflamación/fisiopatología , Dolor/fisiopatología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Receptores Inmunológicos/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Conducta Animal/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Adyuvante de Freund , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/psicología , Inflamación/inducido químicamente , Inyecciones Espinales , Masculino , Dolor/inducido químicamente , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Ratas , Ratas Sprague-Dawley , Receptores Inmunológicos/genética , Receptores de N-Metil-D-Aspartato/genética , Transducción de Señal
17.
Br J Cancer ; 113(9): 1313-22, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26528707

RESUMEN

BACKGROUND: Bone metastases (BMs) are associated with poor outcome in metastatic clear-cell renal carcinoma (m-ccRCC) treated with anti-vascular endothelial growth factor tyrosine kinase inhibitors (anti-VEGFR-TKIs). We aimed to investigate whether expression in the primary tumour of genes involved in the development of BM is associated with outcome in m-ccRCC patients treated with anti-VEGFR-TKIs. METHODS: Metastatic clear-cell renal cell carcinoma patients with available fresh-frozen tumour and treated with anti-VEGFR-TKIs. Quantitative real-time PCR (qRT-PCR) for receptor activator of NF-kB (RANK), RANK-ligand (RANKL), osteoprotegerin (OPG), the proto-oncogene SRC and DKK1 (Dickkopf WNT signalling pathway inhibitor-1). Time-to-event analysis by Kaplan-Meier estimates and Cox regression. RESULTS: We included 129 m-ccRCC patients treated between 2005 and 2013. An elevated RANK/OPG ratio was associated with shorter median time to metastasis (HR 0.50 (95% CI 0.29-0.87); P=0.014), shorter time to BM (HR 0.54 (95% CI 0.31-0.97); P=0.037), shorter median overall survival (mOS) since initial diagnosis (HR 2.27 (95% CI 1.44-3.60); P=0.0001), shorter median progression-free survival (HR 0.44 (95% CI 0.28-0.71); P=0.001) and mOS (HR 0.31 (95% CI 0.19-0.52); P<0.0001) on first-line anti-VEGFR-TKIs in the metastatic setting. Higher RANK expression was associated with shorter mOS on first-line anti-VEGFR-TKIs (HR 0.46 (95% CI 0.29-0.73); P=0.001). CONCLUSIONS: RANK/OPG ratio of expression in primary ccRCC is associated with BM and prognosis in patients treated with anti-VEGFR-TKIs. Prospective validation is warranted.


Asunto(s)
Neoplasias Óseas/patología , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Osteoprotegerina/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/genética , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Neoplasias Óseas/genética , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Supervivencia sin Enfermedad , Femenino , Genes src/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Pronóstico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proto-Oncogenes Mas , Ligando RANK/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
18.
Oncol Rep ; 33(5): 2495-503, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25779224

RESUMEN

Human monopolar spindle-one-binder 2 (hMOB2) is a member of the hMOB family of proteins, and it has been reported to regulate the nuclear-Dbf2-related kinase (NDR) activation. However, the function of hMOB2 expression in tumor cell adhesion and motility has not been addressed. Herein, the lentiviral-mediated overexpression and the knockdown of hMOB2 in HepG2 and SMMC-7721 cells was established. It was demonstrated that overexpression of hMOB2 significantly reduced the cell motility and enhanced the cell-matrix adhesion, while the hMOB2 knockdown decreased not only the cell motility, but also the cell-matrix adhesion. Immunofluorescence results showed that both hMOB2 overexpression and knockdown altered assembly of the focal adhesions and the actin cytoskeleton rearrangement. Furthermore, the focal adhesion kinase (FAK)-Src-paxillin signal pathway activated by hMOB2 was confirmed to be involved in controlling the cell motility and the cell-matrix adhesion. These results demonstrated that the altered cell-matrix adhesion and cell motility induced by hMOB2 expression was caused by the assembly of focal adhesions as well as the actin cytoskeleton rearrangement through the activation of the FAK-Src-paxillin signal pathway, unveiling a novel mechanism of cell motility and cell-matrix adhesion regulation induced by hMOB2 expression.


Asunto(s)
Adhesión Celular/genética , Movimiento Celular/genética , Adhesiones Focales/genética , Invasividad Neoplásica/genética , Proteínas del Tejido Nervioso/genética , Citoesqueleto de Actina/genética , Adhesión Celular/fisiología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/fisiología , Activación Enzimática/genética , Matriz Extracelular/genética , Matriz Extracelular/patología , Quinasa 1 de Adhesión Focal/genética , Adhesiones Focales/patología , Genes src/genética , Células HEK293 , Células Hep G2 , Humanos , Invasividad Neoplásica/patología , Paxillin/genética , Transducción de Señal/genética
19.
Endocrine ; 48(1): 309-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24853881

RESUMEN

To characterize the molecular mechanism and map the response element used by progesterone (P) to upregulate tissue factor (TF) in breast cancer cells. TF expression and mRNA levels were analyzed in breast cancer ZR-75 and T47D cells, using Western blot and real-time PCR, respectively. Mapping of the TF promoter was performed using luciferase vectors. Progesterone receptor (PR) and specificity protein 1 (Sp1) binding to the TF promoter were analyzed by chromatin immuno precipitation assay. Specific or selective inhibitors were used for the MEK1/2 and the c-Src pathways (UO126 and PP2, respectively). TF mRNA increase peaks at 18 h following P treatment in ZR-75 and T47D cells. P upregulation occurs via a transcriptional mechanism that depends on PR and MEK1/2 activation, PR and Sp1 transcription factors bind to a region in the TF promoter that contains three Sp1 sites. TF mRNA upregulation requires an intact PR proline-rich site (mPRO), but it is independent from c-Src. TF upregulation by P is mediated by Sp1 sites in the TF promoter region. Transcriptional upregulation in breast cancer cells occurs via a new mechanism that requires MEK1/2 activation and the mPRO site but independent of c-Src activity. PR Phosphorylation at serine 294 and 345 is not essential.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Progesterona/fisiología , Prolina/fisiología , Receptores de Progesterona/fisiología , Tromboplastina/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genes src/genética , Humanos , Fosforilación , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Regulación hacia Arriba/fisiología
20.
Cell Physiol Biochem ; 34(4): 1402-12, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25301365

RESUMEN

BACKGROUND/AIMS: Functional membrane androgen receptors (mARs) have recently been described in colon tumor cells and tissues. Their activation by specific testosterone albumin conjugates (TAC) down-regulates the PI-3K/Akt pro-survival signaling and triggers potent pro-apoptotic responses both, in vitro and in vivo. The present study explored the mAR-induced regulation of gene products implicated in the tumorigenic activity of Caco2 colon cancer cells. METHODS: In Caco2 human colon cancer cells transcript levels were determined by RT-PCR, protein abundance and phosphorylation by Western blotting and confocal microscopy, as well as cytoskeletal architecture by confocal microscopy. RESULTS: We report time dependent significant decrease in Tyr-416 phosphorylation of c-Src upon mAR activation. In line with the reported late down-regulation of the PI-3K/Akt pathway in testosterone-treated colon tumors, GSK-3beta was phosphorylated at Tyr-216 after long term stimulation of the cells with TAC, a finding supporting the role of this kinase to promote apoptosis. PCR analysis revealed significant decrease of beta-catenin and cyclin D1 transcript levels following TAC treatment. Moreover, confocal laser scanning microscopic analysis disclosed co-localization of beta-catenin with actin cytoskeleton. It is thus conceivable that beta-catenin may participate in the reported modulation of cytoskeletal dynamics in mAR stimulated Caco2 cells. CONCLUSIONS: Our results provide strong evidence that mAR activation regulates late expression and/or activity of the tumorigenic gene products c-Src, GSK-3beta, and beta-catenin thus facilitating the pro-apoptotic response in colon tumor cells.


Asunto(s)
Neoplasias del Colon/genética , Regulación hacia Abajo/genética , Genes src/genética , Glucógeno Sintasa Quinasa 3/genética , Fosforilación/genética , Receptores Androgénicos/metabolismo , beta Catenina/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Albúminas/genética , Albúminas/metabolismo , Apoptosis/genética , Células CACO-2 , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Androgénicos/genética , Transducción de Señal/genética , Testosterona/genética , Testosterona/metabolismo , Transcripción Genética/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...