Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
2.
J Natl Cancer Inst ; 100(2): 109-20, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18182619

ABSTRACT

BACKGROUND: Neuropilin-2 (NRP2) is a high-affinity kinase-deficient receptor for vascular endothelial growth factor (VEGF) and semaphorin 3F. We investigated its function in human colorectal cancers. METHODS: Immunohistochemistry and immunoblotting were used to assess NRP2 expression levels in colorectal tumors and colorectal cancer cell lines, respectively. HCT-116 colorectal cancer cells stably transfected with short hairpin RNA (shRNAs) against NRP2 or control shRNAs were assayed for proliferation by the tetrazolium salt 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and for activation of the VEGFR1 pathway by immunoblotting. Soft agar assays, Annexin V staining, and Boyden chamber assays were used to examine anchorage-independent growth, apoptosis in response to hypoxia, and cell migration/invasion, respectively, in HCT-116 transfectants. Tumor growth and metastasis were analyzed in mice (groups of 10) injected with shRNA-expressing HCT-116 cells. The effect of in vivo targeting of NRP2 by small interfering RNA (siRNA) on the growth of hepatic colorectal tumors derived from luciferase-expressing HCT-116 cells was assessed by measuring changes in bioluminescence and final tumor volumes. All statistical tests were two-sided. RESULTS: NRP2 expression was substantially higher in tumors than in adjacent mucosa. HCT-116 transfectants with reduced NRP2 levels had reduced VEGFR1 signaling, but proliferation was unchanged. Anchorage-independent growth, survival under hypoxic conditions, and motility/invasiveness were also reduced. In vivo, HCT-116 transfectants with reduced NRP2 demonstrated decreased tumor growth, fewer metastases, and increased apoptosis compared with control cells. Hepatic colorectal tumors in mice treated with NRP2 siRNAs were statistically significantly smaller than those in mice treated with control siRNAs (at 28 days after implantation, mean control siRNAs = 420 mm3, mean NRP2 siRNAs = 36 mm3, NRP2 vs control: difference = 385 mm3, 95% confidence interval = 174 mm3 to 595 mm3, P = .005). CONCLUSION: NRP2 on colorectal carcinoma cells is important for tumor growth and is a potential therapeutic target in human cancers where it is expressed.


Subject(s)
Colorectal Neoplasms/metabolism , Liver Neoplasms, Experimental/metabolism , Neuropilin-2/metabolism , RNA, Small Interfering/pharmacology , Animals , Apoptosis/drug effects , Carcinoma/drug therapy , Carcinoma/metabolism , Cell Hypoxia/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Liposomes , Liver Neoplasms, Experimental/drug therapy , Mice , Neoplasm Invasiveness , Neuropilin-2/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Semaphorins/metabolism , Signal Transduction/drug effects , Transfection , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
3.
Cancer Lett ; 258(2): 291-300, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-17950526

ABSTRACT

Both the insulin-like growth factor-I receptor (IGF-IR) and cyclooxygenase-2 (COX-2) are frequently overexpressed in pancreatic cancer. We hypothesized that IGF-IR is directly involved in induction of COX-2 and sought to investigate signaling pathways mediating this effect. Pancreatic cancer cells (L3.6pl) were stably transfected with a dominant-negative receptor (IGF-IR DN) construct or empty vector (pcDNA). Cells were stimulated with IGF-I to determine activated signaling intermediates and induction of COX-2. Signaling pathways mediating COX-2 induction were identified using signaling inhibitors. IGF-I up-regulated COX-2 selectively via the MAPK/(Erk-1/2) pathway. In addition, IGF-IR DN cells showed a marked decrease in constitutive COX-2 and a blunted response to IGF-I. Similarly, treatment with an anti-IGF-IR antibody effectively inhibited IGF-IR and MAPK/Erk activation and decreased COX-2 in parental cells. In conclusion, activation of IGF-IR mediates COX-2 expression in human pancreatic cancer cells.


Subject(s)
Cyclooxygenase 2/genetics , Insulin-Like Growth Factor I/pharmacology , Receptor, IGF Type 1/physiology , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Receptor Substrate Proteins , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptor, IGF Type 1/genetics , Signal Transduction/drug effects , Transfection
4.
J Clin Invest ; 117(8): 2114-22, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17641778

ABSTRACT

We hypothesized that overexpression of PDGF-BB in colorectal cancer (CRC) and pancreatic cancer cells would result in increased pericyte coverage of ECs in vivo, rendering the tumor vasculature more resistant to antiangiogenic therapy. We stably transfected the cDNA for the PDGF-B into HT-29 human CRC and FG human pancreatic cancer cells. Surprisingly, when HT-29 or FG parental and transfected cells were injected into mice (subcutaneously and orthotopically), we observed marked inhibition of tumor growth in the PDGF-BB-overexpressing clones. In the PDGF-BB-overexpressing tumors, we observed an increase in pericyte coverage of ECs. Treatment of PDGF-BB-overexpressing tumors with imatinib mesylate (PDGFR inhibitor) resulted in increased growth and decreased total pericyte content compared with those in untreated PDGF-BB-overexpressing tumors. In vitro studies demonstrated the ability of VSMCs to inhibit EC proliferation by approximately 50%. These data show that increasing the pericyte content of the tumor microenvironment inhibits the growth of angiogenesis-dependent tumors. Single-agent therapy targeting PDGF receptor must be used with caution in tumors when PDGFR is not the target on the tumor cell itself.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/metabolism , Pericytes/metabolism , Platelet-Derived Growth Factor/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Becaplermin , Benzamides , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA, Complementary , Humans , Imatinib Mesylate , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pericytes/pathology , Piperazines/pharmacology , Piperazines/therapeutic use , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins c-sis , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Transfection
5.
Ann Surg Oncol ; 14(10): 2838-46, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17653802

ABSTRACT

BACKGROUND: Colorectal carcinomas (CRC) express high levels of insulin-like growth factor-I/II (IGF-I/II) and the receptor (IGF-IR). We hypothesized that selective inhibition of IGF-IR would inhibit hepatic growth of human CRC in mice. METHODS: Human CRC cells were treated in vitro with anti-IGF-IR monoclonal antibody (MoAB) with and without oxaliplatin to assess cytotoxicity. The effect of anti-IGF-IR MoAB on IGF-I-induced vascular endothelial growth factor (VEGF) production in human CRC cells was assessed by Northern blot and ELISA. We injected human CRC cells intrahepatically in nude mice, and then administered anti-IGF-IR MoAB with and without oxaliplatin. We delayed treatment in one group until large hepatic tumors were present. We assessed tumors for apoptosis, proliferation, and angiogenesis. RESULTS: Anti-IGF-IR MoAB and oxaliplatin inhibited CRC cell growth in vitro and combination treatment was even more effective. IGF-I stimulation of CRC cells resulted in significant upregulation of VEGF and this was completely inhibited by pretreatment with anti-IGF-IR MoAB. Anti-IGF-IR MoAB significantly inhibited hepatic growth of tumors in mice. Anti-IGF-IR MoAB plus oxaliplatin led to a significantly greater inhibition of tumor growth. Anti-IGF-IR MoAB plus oxaliplatin was just as effective at inhibiting growth of larger, more advanced liver tumors. Anti-IGF-IR MoAB, alone and in combination with oxaliplatin, led to a significant increase in tumor cell apoptosis, and a significant inhibition of tumor cell proliferation and angiogenesis. CONCLUSIONS: These findings suggest that IGF-IR is a potential target for therapy in patients with advanced CRC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/pathology , Liver Neoplasms, Experimental/secondary , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Northern , Enzyme-Linked Immunosorbent Assay , HT29 Cells/drug effects , HT29 Cells/pathology , HT29 Cells/transplantation , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Organoplatinum Compounds/pharmacology , Oxaliplatin , Vascular Endothelial Growth Factor A/metabolism
6.
Mol Cancer Ther ; 5(7): 1676-82, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891453

ABSTRACT

Pancreatic carcinoma cells overexpress the insulin-like growth factor-I (IGF-I) receptor (IGF-IR) and the hepatocyte growth factor (HGF) receptor, c-Met, which are both known to mediate tumor cell migration and invasion. We hypothesized that IGF-IR and c-Met cooperate to induce migration and invasion of human pancreatic carcinoma cells and that IGF-I-mediated migration and invasion depend on c-Met. Migration and invasion assays were done with the human pancreatic cancer cell line L3.6pl treated with PBS, IGF-I, HGF, or IGF-I plus HGF. To determine if c-Met is necessary for IGF-IR-mediated migration and invasion, c-Met was down-regulated in L3.6pl cells via adenoviral infection with a c-Met ribozyme before IGF-I treatment. IGF-I and HGF increased cell migration and invasion. Furthermore, IGF-I plus HGF had a greater than additive effect on cell migration and invasion compared with either growth factor alone. Down-regulation of c-Met nearly completely inhibited IGF-I-mediated migration and invasion. Our findings suggest that IGF-IR and c-Met cooperate to induce migration and invasion of human pancreatic carcinoma cells. Furthermore, c-Met is required for both HGF- and IGF-I-mediated migration and invasion. Elucidation of the signaling pathways that contribute to tumor progression and metastasis should provide a foundation for the development of targeted therapies for pancreatic carcinoma.


Subject(s)
Carcinoma/pathology , Cell Movement , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Receptor, IGF Type 1/metabolism , Carcinoma/metabolism , Cell Movement/drug effects , Hepatocyte Growth Factor/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-met/antagonists & inhibitors
7.
Cancer Res ; 65(17): 7775-81, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140945

ABSTRACT

Pancreatic carcinomas express high levels of urokinase-type plasminogen activator (uPA) and its receptor (uPAR), both of which mediate cell migration and invasion. We investigated the hypotheses that (a) insulin-like growth factor-I (IGF-I)- and hepatocyte growth factor (HGF)-mediated migration and invasion of human pancreatic carcinoma cells require uPA and uPAR function and (b) inhibition of uPAR inhibits tumor growth, retroperitoneal invasion, and hepatic metastasis of human pancreatic carcinomas in mice. Using transwell assays, we investigated the effect of IGF-I and HGF on L3.6pl migration and invasion. We measured the induction of uPA and uPAR following treatment of cells with IGF-I and HGF using immunoprecipitation and Western blot analysis. The importance of uPA and uPAR on L3.6pl cell migration and invasion was studied by inhibiting their activities with amiloride and antibodies before cytokine treatment. In an orthotopic mouse model of human pancreatic carcinoma, we evaluated the effect of anti-uPAR monoclonal antibodies with and without gemcitabine on primary tumor growth, retroperitoneal invasion, and hepatic metastasis. IGF-I and HGF mediated cell migration and invasion in L3.6pl cells. In addition, IGF-I and HGF induced uPA and uPAR expression in L3.6pl cells. In vitro, blockade of uPA and uPAR activity inhibited IGF-I- and HGF-mediated cell migration and invasion. Treatment of mice with anti-uPAR monoclonal antibody significantly decreased pancreatic tumor growth and hepatic metastasis and completely inhibited retroperitoneal invasion. Our study shows the importance of the uPA/uPAR system in pancreatic carcinoma cell migration and invasion. These findings suggest that uPAR is a potential target for therapy in patients with pancreatic cancer.


Subject(s)
Cell Movement/physiology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor, IGF Type 1/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Amiloride/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/drug effects , Hepatocyte Growth Factor/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Liver Neoplasms, Experimental/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Proto-Oncogene Proteins c-met/physiology , Receptor, IGF Type 1/physiology , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/immunology , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/biosynthesis
8.
Oncogene ; 24(16): 2647-53, 2005 Apr 14.
Article in English | MEDLINE | ID: mdl-15735759

ABSTRACT

Vascular endothelial growth factor (VEGF) is associated with tumor angiogenesis and poor prognosis in human colorectal cancer (CRC). VEGF receptor-1 (VEGFR-1 or Flt-1) is a high-affinity receptor for VEGF and is typically considered specific to endothelial cells. Here we report the expression and function of VEGFR-1 in CRC cell lines. VEGFR-1 was expressed in all CRC cell lines studied as determined by RT-PCR, Western blot analysis, FACS, and ELISA. Treatment of the human CRC cell lines HT-29 and SW480 with VEGF-A (a ligand for both VEGFR-1 and -2) or VEGF-B (a ligand specific for VEGFR-1) led to activation of Erk-1/2, SAPK/JNK, and translocation of the p65 subunit of nuclear factor-kappaB into the nucleus. Both VEGF-A and -B led to significant induction of cell motility and invasiveness of CRC cells. Stimulation of cells with VEGF-A or -B also led to larger and more numerous colonies in soft agar. However, activation of VEGFR-1 did not increase CRC cell proliferation. In contrast to the previous paradigm that VEGFRs are not present on tumor cells of epithelial origin, we found that VEGFR-1 is present and functional on CRC cells, and activation by VEGF family ligands can activate processes involved in tumor progression and metastasis.


Subject(s)
Colorectal Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Blotting, Western , Cell Movement/drug effects , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL