Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Pancreas ; 42(5): 795-806, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23271399

ABSTRACT

OBJECTIVES: Although c-Src (Src) has emerged as a potential pancreatic cancer target in preclinical studies, Src inhibitors have not demonstrated a significant therapeutic benefit in clinical trials. The objective of these studies was to examine the effects of combining Src inhibition with inhibition of the protein tyrosine phosphatase SHP-2 in pancreatic cancer cells in vitro and in vivo. METHODS: SHP-2 and Src functions were inhibited by siRNA or small molecule inhibitors. The effects of dual Src/SHP-2 functional inhibition were evaluated by Western blot analysis of downstream signaling pathways; cell biology assays to examine caspase activity, viability, adhesion, migration, and invasion in vitro; and an orthotopic nude mouse model to observe pancreatic tumor formation in vivo. RESULTS: Dual targeting of Src and SHP-2 induces an additive or supra-additive loss of phosphorylation of Akt and ERK-1/2 and corresponding increases in expression of apoptotic markers, relative to targeting either protein individually. Combinatorial inhibition of Src and SHP-2 significantly reduces viability, adhesion, migration, and invasion of pancreatic cancer cells in vitro and tumor formation in vivo, relative to individual Src/SHP-2 inhibition. CONCLUSIONS: These data suggest that the antitumor effects of Src inhibition in pancreatic cancer may be enhanced through simultaneous inhibition of SHP-2.


Subject(s)
Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Dasatinib , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , RNA Interference , Signal Transduction/genetics , Thiazoles/administration & dosage , Thiazoles/pharmacology , Tumor Burden/drug effects , Tumor Burden/genetics , Vanadates/administration & dosage , Vanadates/pharmacology , Xenograft Model Antitumor Assays , src-Family Kinases/genetics , src-Family Kinases/metabolism
2.
Mol Carcinog ; 49(12): 1007-21, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20945416

ABSTRACT

Despite an intense focus on novel therapeutic strategies, pancreatic adenocarcinoma remains one of the deadliest human malignancies. The frequent and rapid mortality associated with pancreatic cancer may be attributed to several factors, including late diagnosis, rapid tumor invasion into surrounding tissues, and formation of distant metastases. Both local invasion and metastasis require disruption of tumor cell contacts with the extracellular matrix. Detachment of normal cells from the extracellular matrix leads to a form of programmed cell death termed anoikis. Pancreatic cancer cells avert anoikis by activation of signaling pathways that allow for adhesion-independent survival. In the present studies, cellular signaling pathways activated in detached pancreatic cancer cells were examined. We demonstrate a rapid and robust activation of Src kinase in detached pancreatic cancer cells, relative to adherent. Src autophosphorylation rapidly returned to baseline levels upon reattachment to tissue culture plastic, in the presence or absence of specific extracellular matrix proteins. Treatment of pancreatic cancer cells with tyrosine phosphatase inhibitors increased steady-state Src autophosphorylation in adherent cells and abrogated the detachment-induced increase in Src autophosphorylation. Src was found to co-immunoprecipitate with the Src homology 2 (SH2) domain containing protein tyrosine phosphatase (SHP-2) in pancreatic cancer cells, suggesting that SHP-2 may participate in regulation of Src autophosphorylation in adherent cells. Src family kinase (SFK) dependent increases in Akt and Jun N-terminal kinase (JNK) phosphorylation were observed in detached cells, indicating the potential for Src-dependent activation of survival and stress pathways in pancreatic cancer cells that have detached from the extracellular matrix.


Subject(s)
Adenocarcinoma/enzymology , Pancreatic Neoplasms/enzymology , src-Family Kinases/metabolism , Anoikis , Cell Adhesion , Cell Line, Tumor , Cell Survival , Enzyme Activation , Extracellular Matrix Proteins/metabolism , Gene Knockdown Techniques , Humans , MAP Kinase Kinase 4/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
3.
Nanomedicine ; 6(6): 738-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20553964

ABSTRACT

In an effort to characterize the interaction of cerium oxide nanoparticles (CNPs) in biological systems, we explored the reactivity of CNPs with the phosphate ester bonds of p-nitrophenylphosphate (pNPP), ATP, o-phospho-l-tyrosine, and DNA. The activity of the bond cleavage for pNPP at pH 7 is calculated to be 0.860 ± 0.010 nmol p-nitrophenol/min/µg CNPs. Interestingly, when CNPs bind to plasmid DNA, no cleavage products are detected. While cerium(IV) complexes generally exhibit the ability to break phosphorus-oxygen bonds, the reactions we report appear to be dependent on the availability of cerium(III) sites, not cerium(IV) sites. We investigated the dephosphorylation mechanism from the first principles and find the reaction proceeds through inversion of the phosphate group similar to an S(N)2 mechanism. The ability of CNPs to interact with phosphate ester bonds of biologically relevant molecules has important implications for their use as potential therapeutics.


Subject(s)
Cerium/chemistry , Nanoparticles/chemistry , Organophosphates/chemistry , Adenosine Triphosphate/chemistry , DNA/chemistry , Hydrolysis , Nitrophenols/chemistry , Organophosphorus Compounds/chemistry , Phosphotyrosine/chemistry
4.
Ann Surg Oncol ; 14(12): 3629-37, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17909916

ABSTRACT

BACKGROUND: Pancreatic cancer is an exceptionally lethal disease with an annual mortality nearly equivalent to its annual incidence. This dismal rate of survival is due to several factors including late presentation with locally advanced, unresectable tumors, early metastatic disease, and rapidly arising chemoresistance. To study the mechanisms of chemoresistance in pancreatic cancer we developed two gemcitabine-resistant pancreatic cancer cell lines. METHODS: Resistant cells were obtained by culturing L3.6pl and AsPC-1 cells in serially increasing concentrations of gemcitabine. Stable cultures were obtained that were 40- to 50-fold increased in resistance relative to parental cells. Immunofluorescent staining was performed to examine changes in beta-catenin and E-cadherin localization. Protein expression was determined by immunoblotting. Migration and invasion were determined by modified Boyden chamber assays. Fluorescence-activated cell sorting (FACS) analyses were performed to examine stem cell markers. RESULTS: Gemcitabine-resistant cells underwent distinct morphological changes, including spindle-shaped morphology, appearance of pseudopodia, and reduced adhesion characteristic of transformed fibroblasts. Gemcitabine-resistant cells were more invasive and migratory. Gemcitabine-resistant cells were increased in vimentin and decreased in E-cadherin expression. Immunofluorescence and immunoblotting revealed increased nuclear localization of total beta-catenin. These alterations are hallmarks of epithelial-to-mesenchymal transition (EMT). Resistant cells were activated in the receptor protein tyrosine kinase, c-Met and increased in expression of the stem cell markers CD (cluster of differentiation)24, CD44, and epithelial-specific antigen (ESA). CONCLUSIONS: Gemcitabine-resistant pancreatic tumor cells are associated with EMT, a more-aggressive and invasive phenotype in numerous solid tumors. The increased phosphorylation of c-Met may also be related to chemoresistance and EMT and presents as an attractive adjunctive chemotherapeutic target in pancreatic cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Pancreatic Neoplasms/drug therapy , Cadherins/metabolism , Cell Adhesion , Cell Movement , Deoxycytidine/pharmacology , Epithelium/drug effects , Epithelium/metabolism , Flow Cytometry , Humans , Hyaluronan Receptors/metabolism , Immunoblotting , Membrane Proteins/metabolism , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Neoplasm Invasiveness , Proto-Oncogene Proteins c-met/metabolism , Tumor Cells, Cultured/drug effects , beta Catenin/metabolism , Gemcitabine
5.
J Clin Invest ; 117(10): 2962-73, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17885682

ABSTRACT

The actin filament-associated protein AFAP-110 is an actin cross-linking protein first identified as a substrate of the viral oncogene v-Src. AFAP-110 regulates actin cytoskeleton integrity but also functions as an adaptor protein that affects crosstalk between Src and PKC. Here we investigated the roles of AFAP-110 in the tumorigenic process of prostate carcinoma. Using immunohistochemistry of human tissue arrays, we found that AFAP-110 was absent or expressed at very low levels in normal prostatic epithelium and benign prostatic hyperplasia but significantly increased in prostate carcinomas. The level of AFAP-110 in carcinomas correlated with the Gleason scores. Downregulation of AFAP-110 in PC3 prostate cancer cells inhibited cell proliferation in vitro and tumorigenicity and growth in orthotopic nude mouse models. Furthermore, downmodulation of AFAP-110 resulted in decreased cell-matrix adhesion and cell migration, defective focal adhesions, and reduced integrin beta1 expression. Reintroduction of avian AFAP-110 or a mutant disabling its interaction with Src restored these properties. However, expression of an AFAP-110 lacking the PKC-interacting domain failed to restore properties of parental cells. Thus, increased expression of AFAP-110 is associated with progressive stages of prostate cancer and is critical for tumorigenic growth, in part by regulating focal contacts in a PKC-dependent mechanism.


Subject(s)
Carcinoma/pathology , Cell Transformation, Neoplastic/metabolism , Focal Adhesions/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Prostatic Neoplasms/pathology , Animals , Carcinoma/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Down-Regulation , Extracellular Matrix/pathology , Focal Adhesions/genetics , Humans , Integrins/metabolism , Male , Mice , Mice, Nude , Microfilament Proteins/genetics , Mutation , Phosphoproteins/genetics , Prostatic Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Up-Regulation
6.
Angiogenesis ; 9(2): 101-10, 2006.
Article in English | MEDLINE | ID: mdl-16871430

ABSTRACT

Human pancreatic tumors often overexpress the angiogenesis-promoting factor Interleukin 8 (IL-8), in part due to overexpression of NF-kappaB, a frequent occurrence in pancreatic adenocarcinoma. In this study, we demonstrate that reducing c-Src kinase activity, through either pharmacologic inhibition or small interfering RNA-targeted reduction of Src expression, significantly decreased IL-8 expression (P < 0.05) without affecting NF-kappaB-mediated transcription, but by decreasing phosphorylation of STAT3. To ascertain whether Src-mediated expression of IL-8 was dependent on STAT3, we used stable clones expressing a dominant-negative isoform of STAT3 that inhibits endogenous STAT3 phosphorylation and subsequent DNA binding and STAT3-mediated gene expression or a constitutively activated isoform of STAT3. IL-8 expression was significantly lower in clones expressing the dominant-negative isoform and significantly increased in clones expressing the activated isoform (P < 0.05 for both). Pharmacologic inhibition of NF-kappaB activity significantly reduced basal IL-8 expression and tumor necrosis factor-induced IL-8 expression (P < 0.05 for both), yet NF-kappaB activity was not dependent on Src. We therefore suggest that Src activation, through phosphorylation of STAT3, and NF-kappaB are all required for expression of IL-8 a critical angiogenic-promoting factor in pancreatic adenocarcinomas.


Subject(s)
Adenocarcinoma/metabolism , Interleukin-8/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , STAT3 Transcription Factor/metabolism , Adenocarcinoma/pathology , Cell Line, Tumor , Genes, Reporter , Humans , Immunohistochemistry , Luciferases/metabolism , Microscopy, Confocal , Pancreatic Neoplasms/pathology , Phosphorylation , RNA, Small Interfering/metabolism
7.
Int J Oncol ; 29(1): 125-38, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16773192

ABSTRACT

GN963 is a tyrosine kinase inhibitor with activity against platelet-derived growth factor receptor (PDGFR) and Src kinases. We determined whether oral administration of GN963, alone or in combination with gemcitabine produces therapy against L3.6pl human pancreatic cancer cells growing orthotopically in nude mice. The optimal biological dosage of oral GN963 was determined to be 100 mg/kg every 48 h. Seven days after injection of L3.6pl cells into the pancreas of nude mice, mice (n=10) were treated with vehicle (control), thrice-weekly oral GN963 (100 mg/kg), twice-weekly intraperitoneal gemcitabine (100 mg/kg), or GN963 plus gemcitabine. Treatment with gemcitabine did not significantly differ from control. In contrast, treatment with GN963 (100 mg/kg) or GN963 plus gemcitabine produced a 52% and 81% decrease in tumor volume, respectively. GN963 plus gemcitabine completely inhibited the incidence of liver metastasis. Administration of GN963 inhibited PDGFR phosphorylation in both tumor and tumor-associated endothelial cells, decreased Src and Akt kinase activity in tumor cells, decreased microvessel density, and decreased tumor cell proliferation, while increasing apoptosis of tumor and tumor-associated endothelial cells. Collectively, these data indicate that targeting PDGFR, Src, and Akt on tumor and tumor-associated endothelial cells may be an effective therapy for human pancreatic carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , src-Family Kinases/metabolism , Administration, Oral , Animals , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Drug Therapy, Combination , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Quinoxalines/administration & dosage , Quinoxalines/therapeutic use , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , Gemcitabine
8.
Mini Rev Med Chem ; 6(6): 681-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16787379

ABSTRACT

Selective inhibitors of the Src family of protein tyrosine kinases have been developed as therapeutic agents for human tumors, some of which are now in various stages of clinical trial. In this review, recently described novel small molecule ATP-competitive Src inhibitors are discussed, with an emphasis on their potential use as therapeutic inhibitors for advanced-stage malignancies.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , src-Family Kinases/antagonists & inhibitors , Humans , Neoplasms/enzymology , src-Family Kinases/chemistry , src-Family Kinases/metabolism
9.
Am J Pathol ; 168(3): 962-72, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16507911

ABSTRACT

The nonreceptor protein tyrosine kinase Src is overexpressed in 70% of pancreatic adenocarcinomas. Here, we describe the effect of molecular and pharmacological down-regulation of Src on incidence, growth, and metastasis of pancreatic tumor cells in an orthotopic model. Src expression in L3.6pl human pancreatic tumor cells was reduced by stable expression of a plasmid encoding small interfering RNA (siRNA) to c-src. In stable siRNA clones, Src expression was reduced >80%, with no change in expression of the related kinases c-Yes and c-Lyn, and proliferation rates were similar in all clones. Phosphorylation of Akt and p44/42 Erk mitogen-activated protein kinase and production of VEGF and IL-8 in culture supernatants were also reduced (P < 0.005). On orthotopic implantation of varying cell numbers into nude mice, tumor incidence was unchanged; however, in the siRNA clones, large tumors failed to develop, and incidence of metastasis was significantly reduced, suggesting that c-Src activity is critical to tumor progression. To examine this possibility further, animals bearing established wild-type tumors were treated with the Src/Abl-selective inhibitor BMS-354825 (dasatinib). Tumor size was decreased, and incidence of metastases was significantly reduced in treated mice compared with controls. These results demonstrate that Src activation contributes to pancreatic tumor progression in this model, offering Src as a candidate for targeted therapy.


Subject(s)
Adenocarcinoma/enzymology , Adenocarcinoma/secondary , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Adenocarcinoma/blood supply , Animals , Cell Line, Tumor , Dasatinib , Disease Models, Animal , Disease Progression , Humans , Interleukin-8/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Pathologic/enzymology , Pancreatic Neoplasms/blood supply , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Thiazoles/pharmacology , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/metabolism
11.
Mol Cancer Ther ; 4(12): 1900-11, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16373705

ABSTRACT

c-Src is frequently activated in human malignancies, including colon, breast, and pancreatic carcinomas. Several recent studies have shown that activation of Src family kinases leads to tumor progression and metastasis by increasing cellular migration and invasion, promoting cell growth and survival, and deregulating expression of proangiogenic molecules. Therefore, selective inhibitors of Src are being developed for cancer therapy. In this study, we characterize the biological effects of the novel ATP-based Src family kinase inhibitor, AP23846, in tumor cells with high Src activity. As a lead compound, AP23846 is a potent c-Src kinase inhibitor (IC50 approximately 0.5 nmol/L in vitro, approximately 10-fold more potent than PP2, the most widely used commercially available Src family kinase inhibitor). At concentrations of 1 micromol/L, AP23846 led to complete Src inhibition for 48 hours in cells. No cytotoxicity was observed under these conditions, although proliferation rates were slower. Therefore, this was an excellent inhibitor to examine Src-regulated signaling pathways in tumor cells. AP23846 reduced cellular migration, vascular endothelial growth factor, and interleukin-8 in a dose-dependent fashion in pancreatic adenocarcinoma cells grown in vitro. Correspondingly, cell culture supernatants from L3.6pl pancreatic adenocarcinoma cells pretreated with AP23846 failed to promote migration of hepatic endothelial cells in vitro and failed to support angiogenesis into gel foams implanted s.c. in mice in vivo. These results suggest that Src inhibitors affect biological properties of tumor progression and may be useful as cancer therapeutic agents in more advanced disease.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Enzyme Inhibitors/pharmacology , Interleukin-8/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/antagonists & inhibitors , Adenosine Triphosphate/pharmacology , Animals , Base Sequence , Blotting, Western , Cell Line, Tumor , Cell Movement , DNA Primers , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunoprecipitation , Mice , Mice, Inbred C3H , Neoplasms/blood supply , Phosphorylation , RNA, Small Interfering
12.
Pancreas ; 31(3): 263-74, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16163059

ABSTRACT

OBJECTIVES: Multiple signaling proteins may be aberrantly activated and/or overexpressed in pancreatic tumors, including the nonreceptor protein tyrosine kinase Src. The goal of this study was to determine the role of Src in regulating VEGF expression and angiogenic potential in pancreatic cancer cell lines. METHODS: Src activity was inhibited using the Src family kinase selective inhibitor PP2, and c-Src expression was down-regulated via siRNA. The activities of downstream signaling molecules phosphatidyl inositol 3'-kinase (PI3K) and p38 mitogen-activated protein kinase (MAPK) were disrupted via selective inhibitors. In vivo angiogenesis was assessed through the use of a gel-foam assay. RESULTS: Inhibition of Src activity or expression decreases both constitutive and EGF-induced VEGF production. Both the PI3K/Akt and p38 MAPK pathways are activated in a Src family kinase-dependent fashion on EGF-R activation and are important for EGF-mediated VEGF production in pancreatic cancer cells. Additionally, media from Src-inhibited L3.6pl cells fail to promote angiogenesis into gel foams implanted subcutaneously into mice, whereas media from control cells promote a robust angiogenic response. CONCLUSIONS: Src activity contributes to constitutive and EGF-induced VEGF expression and angiogenic potential in pancreatic cancer cells. Therefore, Src may be a viable target for antiangiogenesis therapy in pancreatic cancer.


Subject(s)
MAP Kinase Signaling System/physiology , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , Female , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C3H , Pancreatic Neoplasms/blood supply , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
13.
Cancer Res ; 65(16): 7214-22, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103072

ABSTRACT

Interleukin-8 (IL-8) is an angiogenic factor that promotes growth of pancreatic tumors. The purpose of this study was to determine if c-Src, a protein tyrosine kinase frequently overexpressed in pancreatic cancer, regulated IL-8 expression and to elucidate the Src-mediated signaling pathways that contribute to angiogenesis in pancreatic adenocarcinoma cells. In a panel of pancreatic cancer cell lines, expression of total and activated Src correlated with IL-8 production. Furthermore, ectopic expression of activated Src in PANC-1 cells with low endogenous Src activity significantly increased IL-8 production (P < 0.005). In contrast, pharmacologic inhibition of endogenous c-Src kinase activity or small interfering RNA-mediated "knockdown" of c-Src expression in L3.6pl cells with high Src expression and activity caused significant decreases in IL-8 production (P < 0.005). Inhibition of c-Src activity resulted in decreased phosphorylation of Akt, p38, and extracellular signal-regulated kinase (Erk)-1/2. Significant (P < 0.005) dose-dependent decreases were observed in IL-8 expression by inhibiting Src-dependent signaling molecules Erk-1/2 and p38 but not phosphatidylinositol 3-kinase. To assess the relevance of Src inhibition to angiogenesis, in vivo gelfoam assays were done. Robust infiltration of vessels was observed in gelfoam saturated with conditioned medium from pancreatic carcinoma cells. This angiogenesis was nearly abrogated in gelfoams saturated with conditioned medium from cells treated with the Src family kinase inhibitor, PP2 (P < 0.001). Thus, c-Src regulates critical "downstream" signaling pathways that contribute to expression of IL-8 in human pancreatic tumor cells, suggesting c-Src may be a target for therapeutic intervention in pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/blood supply , Interleukin-8/biosynthesis , Pancreatic Neoplasms/blood supply , Phosphotransferases/metabolism , Proto-Oncogene Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Doxycycline/pharmacology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-8/antagonists & inhibitors , MAP Kinase Signaling System , Mice , Mice, Inbred C3H , Mice, Nude , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphorylation , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/biosynthesis , Phosphotransferases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/genetics , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases
14.
Cancer Res ; 65(15): 6493-7, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061624

ABSTRACT

Experimental evidence suggests that CXCR4, a Gi protein-coupled receptor for the ligand CXCL12/stromal cell-derived factor-1alpha (SDF-1alpha), plays a role in breast cancer metastasis. Transactivation of HER2-neu by G protein-coupled receptor activation has been reported as a ligand-independent mechanism of activating tyrosine kinase receptors. We found that SDF-1alpha transactivated HER2-neu in the breast cancer cell lines MDA-MB-361 and SKBR3, which express both CXCR4 and HER2-neu. AMD3100, a CXCR4 inhibitor, PKI 166, an epidermal growth factor receptor/HER2-neu tyrosine kinase inhibitor, and PP2, a Src kinase inhibitor, each blocked SDF-1alpha-induced HER2-neu phosphorylation. Blocking Src kinase, with PP2 or using a kinase-inactive Src construct, and inhibiting epidermal growth factor receptor/HER2-neu signaling with PKI 166 each inhibited SDF-1alpha-stimulated cell migration. We report a novel mechanism of HER2-neu transactivation through SDF-1alpha stimulation of CXCR4 that involves Src kinase activation.


Subject(s)
Breast Neoplasms/metabolism , Chemokines, CXC/physiology , Receptor, ErbB-2/biosynthesis , src-Family Kinases/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , Chemokine CXCL12 , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Enzyme Activation , Humans , Receptor, ErbB-2/genetics , Signal Transduction , Transcriptional Activation , src-Family Kinases/genetics
15.
Clin Cancer Res ; 11(1): 397-405, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15671571

ABSTRACT

Targeted therapies that inhibit the activity of tyrosine kinase receptors such as the epidermal growth factor receptor (EGFR) have shown activity against solid malignancies when used as single agents or in combination with chemotherapy. Although anti-EGFR therapies are active in some patients, eventually disease in nearly all patients will become refractory to therapy. Therefore, a better understanding of the mechanisms of resistance to anti-EGFR therapies is critical to further improve the efficacy of this class of agents. Mechanisms that mediate resistance to anti-EGFR therapies include the presence of redundant tyrosine kinase receptors, increased angiogenesis, and the constitutive activation of downstream mediators. Two recent landmark publications have also shown that specific mutations in the kinase domain of EGFR in some lung carcinomas are associated with markedly improved response rates to an EGFR tyrosine kinase inhibitor. Mutations in the EGFR receptor seem to play a significant role in determining the sensitivity of tumor cells to EGFR inhibitor therapy by altering the conformation and activity of the receptor. As the field of molecular therapeutics continues to evolve, a comprehensive understanding of resistance mechanisms will ultimately lead to refinements in our regimens to provide better care for patients with cancer.


Subject(s)
Carcinoma/pathology , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Lung Neoplasms/genetics , Neovascularization, Pathologic , ErbB Receptors/antagonists & inhibitors , Female , Humans , Ligands , Lung Neoplasms/pathology , Male , Models, Biological , Mutation , Protein Structure, Tertiary , Signal Transduction , Skin Neoplasms/drug therapy , Time Factors , Treatment Outcome
16.
Cancer Metastasis Rev ; 22(4): 337-58, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12884910

ABSTRACT

The Src family of non-receptor protein tyrosine kinases plays critical roles in a variety of cellular signal transduction pathways, regulating such diverse processes as cell division, motility, adhesion, angiogenesis, and survival. Constitutively activated variants of Src family kinases, including the viral oncoproteins v-Src and v-Yes, are capable of inducing malignant transformation of a variety of cell types. Src family kinases, most notably although not exclusively c-Src, are frequently overexpressed and/or aberrantly activated in a variety of epithelial and non-epithelial cancers. Activation is very common in colorectal and breast cancers, and somewhat less frequent in melanomas, ovarian cancer, gastric cancer, head and neck cancers, pancreatic cancer, lung cancer, brain cancers, and blood cancers. Further, the extent of increased Src family activity often correlates with malignant potential and patient survival. Activation of Src family kinases in human cancers may occur through a variety of mechanisms and is frequently a critical event in tumor progression. Exactly how Src family kinases contribute to individual tumors remains to be defined completely, however they appear to be important for multiple aspects of tumor progression, including proliferation, disruption of cell/cell contacts, migration, invasiveness, resistance to apoptosis, and angiogenesis. This review details the evidence for Src family activation in human tumors, and emphasizes possible consequences to tumor progression. Given the ability of Src and its family members to participate in so many aspects of tumor progression and metastasis, Src family kinases are attractive targets for future anti-cancer therapeutics.


Subject(s)
Neoplasms/enzymology , Neoplasms/pathology , Disease Progression , Enzyme Activation , Humans , Neoplasm Metastasis , Neovascularization, Pathologic , Signal Transduction , src-Family Kinases/metabolism
17.
J Cell Sci ; 116(Pt 12): 2585-98, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12734402

ABSTRACT

c-Src and c-Yes are highly homologous members of the Src family of non-receptor tyrosine kinases. The overall sequence similarity between c-Src and c-Yes allows them to perform many overlapping functions. However, the phenotypes of the c-src and c-yes knockout mice, and cells derived from them, are quite different, indicating functional specificity between the two proteins. Specifically, c-src-/- cells are deficient in several processes that require dynamic regulation of the actin cytoskeleton. In order to begin to understand why c-Yes is unable to compensate for c-Src signaling, we used a series of Src/Yes chimeras in which the non-catalytic functional domains of Src527F were replaced by those of c-Yes. Using chicken embryo fibroblasts as a model system, our results indicate that the c-Yes N-terminal SH4-Unique domains are sufficient to inhibit the ability of Src527F to alter cell morphology, induce actin filament rearrangements or stimulate motility or invasive potential. The data also indicate that the SH4-Unique-SH3-SH2 domains of c-Yes work cooperatively and prevent activation of signaling proteins associated with Src527F transformation, including activation of phosphatidylinositol 3-kinase, phosphorylation of c-Raf and Akt and downregulation of RhoA-GTP. These data indicate that c-Yes may not modulate signals associated with c-Src-induced changes in actin filament integrity and may explain why c-Yes fails to compensate for c-Src signaling in src-/- cells.


Subject(s)
Actin Cytoskeleton/metabolism , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/deficiency , Proto-Oncogene Proteins/metabolism , Signal Transduction/genetics , src Homology Domains/physiology , src-Family Kinases , Animals , CSK Tyrosine-Protein Kinase , Cell Movement/genetics , Cell Size/genetics , Cells, Cultured , Chick Embryo , Fibroblasts/cytology , Fibroblasts/enzymology , Gene Expression Regulation, Enzymologic/genetics , MAP Kinase Signaling System/genetics , Neoplasm Invasiveness/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-yes , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
18.
Front Biosci ; 8: s185-205, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12456296

ABSTRACT

c-Yes and c-Src are the two most closely related members of the Src family of nonreceptor tyrosine kinases. Although there is much evidence to support redundancy in signaling between these two kinases, there is also a growing body of evidence to indicate specificity in signaling. In this review, we summarize c-Yes, its potential functions and its ability to modulate signals that are distinct from c-Src.


Subject(s)
Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , src-Family Kinases , Animals , Humans , Proto-Oncogene Proteins c-yes , Substrate Specificity
19.
Oncogene ; 21(51): 7797-807, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12420216

ABSTRACT

Src is a non-receptor protein tyrosine kinase, the expression and activity of which is increased in >80% of human colon cancers with respect to normal colonic epithelium. Previous studies from this and other laboratories have demonstrated that Src activity contributes to tumorigenicity of established colon adenocarcinoma cell lines. Src participates in the regulation of many signal transduction pathways, among which are those leading to cellular survival. In this study, we addressed the potential role of Src activation to a specific aspect of tumor cell survival, resistance to detachment-induced apoptosis (anoikis). Using five colon tumor cell lines with different biologic properties and genetic alterations, we demonstrate that expression and activity of Src corresponds with resistance to anoikis. Enforced expression of activated Src in subclones of SW480 cells (of low intrinsic Src expression and activity) increases resistance to anoikis; whereas decreased Src expression in HT29 cells (of high Src expression and activity) by transfection with anti-sense Src expression vectors increases susceptibility to anoikis. In contrast, increasing or decreasing Src expression had no effect on susceptibility to staurosporine-induced apoptosis in attached cells. PD173955, a Src family-specific tyrosine kinase inhibitor, increases the susceptibility of HT29 cells to anoikis in a dose- and time-dependent manner. Increasing Src expression and activity led to increased phosphorylation of Akt, a mediator of cellular survival pathways, whereas decreasing Src activity led to decreased Akt phosphorylation. In colon tumor cells with high Src activity, the PI3 kinase inhibitor LY 294002 sensitized cells to anoikis. These results suggest that Src activation may contribute to colon tumor progression and metastasis in part by activating Akt-mediated survival pathways that decrease sensitivity of detached cells to anoikis.


Subject(s)
Adenocarcinoma/pathology , Anoikis/physiology , Colonic Neoplasms/pathology , Neoplasm Proteins/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins pp60(c-src)/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Amino Acid Substitution , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Adhesion , Cell Cycle , Chromones/pharmacology , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Disease Progression , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Genes, src , Humans , Morpholines/pharmacology , Mutation, Missense , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidines/pharmacology , Recombinant Fusion Proteins/physiology , Staurosporine/pharmacology , Transfection , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , bcl-X Protein
20.
Mol Biol Cell ; 13(7): 2311-22, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12134071

ABSTRACT

The actin filament-associated protein and Src-binding partner, AFAP-110, is an adaptor protein that links signaling molecules to actin filaments. AFAP-110 binds actin filaments directly and multimerizes through a leucine zipper motif. Cellular signals downstream of Src(527F) can regulate multimerization. Here, we determined recombinant AFAP-110 (rAFAP-110)-bound actin filaments cooperatively, through a lateral association. We demonstrate rAFAP-110 has the capability to cross-link actin filaments, and this ability is dependent on the integrity of the carboxy terminal actin binding domain. Deletion of the leucine zipper motif or PKC phosphorylation affected AFAP-110's conformation, which correlated with changes in multimerization and increased the capability of rAFAP-110 to cross-link actin filaments. AFAP-110 is both a substrate and binding partner of PKC. On PKC activation, stress filament organization is lost, motility structures form, and AFAP-110 colocalizes strongly with motility structures. Expression of a deletion mutant of AFAP-110 that is unable to bind PKC blocked the effect of PMA on actin filaments. We hypothesize that upon PKC activation, AFAP-110 can be cooperatively recruited to newly forming actin filaments, like those that exist in cell motility structures, and that PKC phosphorylation effects a conformational change that may enable AFAP-110 to promote actin filament cross-linking at the cell membrane.


Subject(s)
Actins/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Actinin/metabolism , Animals , Binding Sites , Cell Line , Cross-Linking Reagents/metabolism , Enzyme Activation , Leucine Zippers , Microfilament Proteins/genetics , Phosphoproteins/genetics , Phosphorylation , Polymers/metabolism , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...