Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Theranostics ; 11(16): 8092-8111, 2021.
Article in English | MEDLINE | ID: mdl-34335982

ABSTRACT

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.


Subject(s)
CSK Tyrosine-Protein Kinase/metabolism , Carcinoma, Hepatocellular/metabolism , Membrane Proteins/metabolism , CSK Tyrosine-Protein Kinase/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/physiology , Genes, src/genetics , Genes, src/physiology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/physiology , Peptides/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tetraspanins/genetics , Tetraspanins/metabolism
2.
Ann Med ; 53(1): 596-610, 2021 12.
Article in English | MEDLINE | ID: mdl-33830879

ABSTRACT

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.


Subject(s)
Adaptive Immunity/genetics , Genes, src/genetics , Genomics/statistics & numerical data , Immunotherapy , Urinary Bladder Neoplasms/genetics , Annexin A1/metabolism , Area Under Curve , Beclin-1/metabolism , Biomarkers, Tumor/genetics , Databases, Genetic , ErbB Receptors/metabolism , Gene Expression/genetics , Genomics/methods , Humans , Machine Learning , Patient Selection , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Risk Factors , Survival Analysis , Urinary Bladder Neoplasms/drug therapy , Axl Receptor Tyrosine Kinase
3.
J Neurosci ; 41(17): 3808-3821, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33727334

ABSTRACT

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.


Subject(s)
Axons/physiology , Paxillin/physiology , Spinal Cord/growth & development , Animals , Axon Guidance/physiology , Chick Embryo , Electroporation , Ephrins/physiology , Female , Gene Knockdown Techniques , Genes, src/genetics , Humans , Male , Mice , MicroRNAs/genetics , Motor Neurons/physiology , Mutation/genetics , Neurites/physiology , Spinal Cord/cytology
4.
Sci Rep ; 9(1): 4819, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894630

ABSTRACT

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.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Epithelial-Mesenchymal Transition/genetics , Genes, src/genetics , Lung Neoplasms/genetics , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Culture Techniques , Cell Line, Tumor , Extracellular Matrix/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/pathology , Mice , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Rats , Signal Transduction/genetics , Spheroids, Cellular/pathology , Tumor Microenvironment/genetics
5.
Int J Mol Sci ; 20(5)2019 Mar 06.
Article in English | MEDLINE | ID: mdl-30845661

ABSTRACT

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.


Subject(s)
12E7 Antigen/genetics , 12E7 Antigen/metabolism , Brain Neoplasms/genetics , Gene Expression Profiling/methods , Glioblastoma/genetics , Up-Regulation , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genes, src/genetics , Glioblastoma/metabolism , Humans , Neoplasm Invasiveness , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/pharmacology , Sequence Analysis, RNA
6.
Oncogene ; 38(21): 4015-4027, 2019 05.
Article in English | MEDLINE | ID: mdl-30692633

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Insulin-Like Growth Factor I/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Genes, src/genetics , Humans , MCF-7 Cells , Methylation , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/physiology , Receptor, IGF Type 1/metabolism
7.
Cell Death Dis ; 9(9): 877, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30158525

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Genes, src/genetics , Proto-Oncogene Proteins p21(ras)/genetics , src-Family Kinases/genetics , Apoptosis/genetics , Caco-2 Cells , Cell Line, Tumor , ErbB Receptors/genetics , Gefitinib/pharmacology , HCT116 Cells , HT29 Cells , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Quinazolines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Cancer Res ; 78(11): 2825-2838, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29531159

ABSTRACT

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.


Subject(s)
Carcinogenesis/genetics , Genes, src/genetics , Protein Processing, Post-Translational/genetics , STAT3 Transcription Factor/genetics , Acetylation , Animals , CREB-Binding Protein/genetics , CSK Tyrosine-Protein Kinase , Cell Line , Cell Line, Tumor , Cell Nucleus/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Nuclear Proteins/genetics , Phosphorylation/genetics , Protein-Tyrosine Kinases/genetics , Trans-Activators/genetics , Transcription, Genetic/genetics , src-Family Kinases/genetics
9.
Neoplasia ; 20(3): 233-243, 2018 03.
Article in English | MEDLINE | ID: mdl-29444487

ABSTRACT

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.


Subject(s)
Cell Transformation, Neoplastic/pathology , Fibroblast Growth Factors/genetics , Genes, src/genetics , Intercellular Signaling Peptides and Proteins/genetics , Oncogenes/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line , Cell Transformation, Neoplastic/genetics , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Stromal Cells/metabolism , Stromal Cells/pathology
10.
J BUON ; 22(5): 1097-1106, 2017.
Article in English | MEDLINE | ID: mdl-29135089

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/complications , Colorectal Neoplasms/genetics , Focal Adhesion Kinase 1/metabolism , Genes, src/genetics , Liver Neoplasms/secondary , Paxillin/metabolism , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Liver Neoplasms/pathology , Male , Middle Aged
11.
J Pharmacol Exp Ther ; 363(3): 428-443, 2017 12.
Article in English | MEDLINE | ID: mdl-29021381

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Focal Adhesion Protein-Tyrosine Kinases/genetics , Genes, src/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , A549 Cells , Animals , Dasatinib/pharmacology , Drug Synergism , Enzyme Activation , Humans , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Nude , Niacinamide/pharmacology , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Sorafenib , Xenograft Model Antitumor Assays
12.
Mediators Inflamm ; 2017: 8123281, 2017.
Article in English | MEDLINE | ID: mdl-28804222

ABSTRACT

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.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Epithelial-Mesenchymal Transition/drug effects , Genes, src/physiology , Transforming Growth Factor beta1/pharmacology , A549 Cells , Cell Movement/drug effects , Endoplasmic Reticulum Chaperone BiP , Genes, src/genetics , Humans , Nasal Polyps/metabolism , Organ Culture Techniques , Signal Transduction/drug effects
13.
J Endocrinol ; 233(2): 175-186, 2017 05.
Article in English | MEDLINE | ID: mdl-28223314

ABSTRACT

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.


Subject(s)
Epidermal Growth Factor/pharmacology , Gene Expression Regulation/physiology , Growth Hormone/metabolism , Signal Transduction/physiology , Animals , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, src/genetics , Genes, src/physiology , Growth Hormone/genetics , Humans , Liver/metabolism , Mice , Mice, Transgenic , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Mol Biol Cell ; 27(24): 3926-3936, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27733622

ABSTRACT

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.


Subject(s)
Genes, src/genetics , Genes, src/physiology , Proto-Oncogene Proteins c-fyn/metabolism , Acylation , Amino Acid Sequence , Animals , COS Cells , CSK Tyrosine-Protein Kinase , Caveolin 1/metabolism , Cell Membrane/metabolism , Chlorocebus aethiops , Cysteine/metabolism , Green Fluorescent Proteins , Lipoylation , Membrane Proteins , Membranes/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
15.
Avian Pathol ; 45(2): 202-7, 2016.
Article in English | MEDLINE | ID: mdl-27100152

ABSTRACT

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.


Subject(s)
Avian Leukosis Virus/genetics , Avian Leukosis/virology , Chickens/virology , Chromosome Aberrations , Fibrosarcoma/veterinary , Genes, src/genetics , Acute Disease , Animals , Avian Leukosis/genetics , Avian Leukosis Virus/isolation & purification , Chick Embryo , Female , Fibrosarcoma/genetics , Fibrosarcoma/virology , Karyotype , Karyotyping/veterinary , Polymorphism, Genetic
16.
Anesth Analg ; 122(3): 871-881, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26720617

ABSTRACT

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.


Subject(s)
Genes, src/genetics , Inflammation/physiopathology , Pain/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Behavior, Animal/drug effects , Enzyme Inhibitors/pharmacology , Freund's Adjuvant , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/psychology , Inflammation/chemically induced , Injections, Spinal , Male , Pain/chemically induced , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction
17.
Br J Cancer ; 113(9): 1313-22, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26528707

ABSTRACT

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.


Subject(s)
Bone Neoplasms/pathology , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Osteoprotegerin/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Bone Neoplasms/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Disease-Free Survival , Female , Genes, src/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Male , Middle Aged , Prognosis , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Mas , RANK Ligand/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Oncol Rep ; 33(5): 2495-503, 2015 May.
Article in English | MEDLINE | ID: mdl-25779224

ABSTRACT

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.


Subject(s)
Cell Adhesion/genetics , Cell Movement/genetics , Focal Adhesions/genetics , Neoplasm Invasiveness/genetics , Nerve Tissue Proteins/genetics , Actin Cytoskeleton/genetics , Cell Adhesion/physiology , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Enzyme Activation/genetics , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Focal Adhesion Kinase 1/genetics , Focal Adhesions/pathology , Genes, src/genetics , HEK293 Cells , Hep G2 Cells , Humans , Neoplasm Invasiveness/pathology , Paxillin/genetics , Signal Transduction/genetics
19.
Endocrine ; 48(1): 309-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24853881

ABSTRACT

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.


Subject(s)
MAP Kinase Signaling System/physiology , Progesterone/physiology , Proline/physiology , Receptors, Progesterone/physiology , Thromboplastin/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation/drug effects , Genes, src/genetics , Humans , Phosphorylation , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Up-Regulation/physiology
20.
Cell Physiol Biochem ; 34(4): 1402-12, 2014.
Article in English | MEDLINE | ID: mdl-25301365

ABSTRACT

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.


Subject(s)
Colonic Neoplasms/genetics , Down-Regulation/genetics , Genes, src/genetics , Glycogen Synthase Kinase 3/genetics , Phosphorylation/genetics , Receptors, Androgen/metabolism , beta Catenin/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Albumins/genetics , Albumins/metabolism , Apoptosis/genetics , Caco-2 Cells , Cell Line, Tumor , Colonic Neoplasms/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/genetics , Signal Transduction/genetics , Testosterone/genetics , Testosterone/metabolism , Transcription, Genetic/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...