Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
BMC Cancer ; 13: 321, 2013 Jul 01.
Article En | MEDLINE | ID: mdl-23815882

BACKGROUND: Human mitochondrial peptide deformylase (PDF) has been proposed as a novel cancer therapeutic target. However, very little is known about its expression and regulation in human tissues. The purpose of this study was to characterize the expression pattern of PDF in cancerous tissues and to identify mechanisms that regulate its expression. METHODS: The mRNA expression levels of PDF and methionine aminopeptidase 1D (MAP1D), an enzyme involved in a related pathway with PDF, were determined using tissue panels containing cDNA from patients with various types of cancer (breast, colon, kidney, liver, lung, ovarian, prostate, or thyroid) and human cell lines. Protein levels of PDF were also determined in 2 colon cancer patients via western blotting. Colon cancer cells were treated with inhibitors of ERK, Akt, and mTOR signaling pathways and the resulting effects on PDF and MAP1D mRNA levels were determined by qPCR for colon and lung cancer cell lines. Finally, the effects of a PDF inhibitor, actinonin, on the proliferation of breast, colon, and prostate cell lines were determined using the CyQUANT assay. RESULTS: PDF and MAP1D mRNA levels were elevated in cancer cell lines compared to non-cancer lines. PDF mRNA levels were significantly increased in breast, colon, and lung cancer samples while MAP1D mRNA levels were increased in just colon cancers. The expression of PDF and MAP1D varied with stage in these cancers. Further, PDF protein expression was elevated in colon cancer tissue samples. Inhibition of the MEK/ERK, but not PI3K or mTOR, pathway reduced the expression of PDF and MAP1D in both colon and lung cancer cell lines. Further, inhibition of PDF with actinonin resulted in greater reduction of breast, colon, and prostate cancer cell proliferation than non-cancer cell lines. CONCLUSIONS: This is the first report showing that PDF is over-expressed in breast, colon, and lung cancers, and the first evidence that the MEK/ERK pathway plays a role in regulating the expression of PDF and MAP1D. The over-expression of PDF in several cancers and the inhibition of cancer cell growth by a PDF inhibitor suggest this enzyme may act as an oncogene to promote cancer cell proliferation.


Amidohydrolases/biosynthesis , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , Lung Neoplasms/metabolism , Amidohydrolases/analysis , Aminopeptidases/analysis , Aminopeptidases/biosynthesis , Blotting, Western , Female , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Tissue Array Analysis
2.
Cell Signal ; 25(5): 1156-65, 2013 May.
Article En | MEDLINE | ID: mdl-23416466

MAP kinase (MAPK) signaling results from activation of Raf kinases in response to external or internal stimuli. Here, we demonstrate that Raf kinase inhibitory protein (RKIP) regulates the activation of MAPK when B-Raf signaling is defective. We used multiple models including mouse embryonic fibroblasts (MEFs) and primary keratinocytes from RKIP- or Raf-deficient mice as well as allografts in mice to investigate the mechanism. Loss of B-Raf protein or activity significantly reduces MAPK activation in these cells. We show that RKIP depletion can rescue the compromised ERK activation and promote proliferation, and this rescue occurs through a Raf-1 dependent mechanism. These results provide formal evidence that RKIP is a bona fide regulator of Raf-1. We propose a new model in which RKIP plays a key role in regulating the ability of cells to signal through Raf-1 to ERK in B-Raf compromised cells.


Mitogen-Activated Protein Kinases/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Animals , Cell Proliferation , Cells, Cultured , Enzyme Activation , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylethanolamine Binding Protein/antagonists & inhibitors , Phosphatidylethanolamine Binding Protein/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction
3.
PLoS One ; 7(11): e49893, 2012.
Article En | MEDLINE | ID: mdl-23166790

Angiogenesis inhibition is an important therapeutic strategy for advanced stage prostate cancer. Previous work from our laboratory showed that sustained stimulation of Rap1 by 8-pCPT-2'-O-Me-cAMP (8CPT) via activation of Epac, a Rap1 GEF, or by expression of a constitutively active Rap1 mutant (cRap1) suppresses endothelial cell chemotaxis and subsequent angiogenesis. When we tested this model in the context of a prostate tumor xenograft, we found that 8CPT had no significant effect on prostate tumor growth alone. However, in cells harboring cRap1, 8CPT dramatically inhibited not only prostate tumor growth but also VEGF expression and angiogenesis within the tumor microenvironment. Subsequent analysis of the mechanism revealed that, in prostate tumor epithelial cells, 8CPT acted via stimulation of PKA rather than Epac/Rap1. PKA antagonizes Rap1 and hypoxic induction of 1α protein expression, VEGF production and, ultimately, angiogenesis. Together these findings provide evidence for a novel interplay between Rap1, Epac, and PKA that regulates tumor-stromal induction of angiogenesis.


Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/analogs & derivatives , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/physiopathology , rap1 GTP-Binding Proteins/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Line, Tumor , Cyclic AMP/pharmacology , Enzyme-Linked Immunosorbent Assay , Guanine Nucleotide Exchange Factors/metabolism , Humans , Immunohistochemistry , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Prostatic Neoplasms/drug therapy , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors
4.
Curr Cancer Drug Targets ; 11(4): 394-404, 2011 May.
Article En | MEDLINE | ID: mdl-21395552

Lung cancer is a leading cause of death in both men and women, with over 1,000,000 new cases diagnosed worldwide annually and a 5-year survival rate of only 14%, a figure that has improved little in the past thirty years. This poor prognosis suggests a need for novel approaches for the treatment and prevention of lung cancer. The renin-angiotensin system is an established, primary regulator of blood pressure, homeostasis, and natriuresis; however, compelling evidence indicates that the angiotensin peptides also play a role in cell proliferation and inflammation. Angiotensin II is a vasoconstrictor, a mitogen, and an angiogenic factor, while angiotensin-(1-7) has vasodilator, anti-proliferative, and anti-angiogenic properties. This review focuses on studies examining the renin-angiotensin system in pulmonary cancers and whether clinical intervention of this pathway may serve as an effective chemotherapeutic and/or chemopreventive modality for lung cancer.


Angiotensins/metabolism , Angiotensins/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Animals , Humans , Peptide Fragments/metabolism , Peptide Fragments/pharmacology
5.
Mol Cancer Ther ; 8(6): 1676-83, 2009 Jun.
Article En | MEDLINE | ID: mdl-19509262

Angiotensin-(1-7) [Ang-(1-7)] is an endogenous seven-amino acid peptide hormone with antiproliferative properties. Our previous studies showed that Ang-(1-7) inhibits the growth of human lung cancer cells in vitro and reduces the size of human lung tumor xenografts in vivo. In the current study, s.c. injection of Ang-(1-7) not only caused a significant reduction in human A549 lung tumor growth but also markedly decreased vessel density, suggesting that the heptapeptide inhibits angiogenesis to reduce tumor size. A decrease in human endothelial cell tubule formation in Matrigel was observed following a 16 h incubation with Ang-(1-7), with a maximal reduction at a 10 nmol/L concentration. Ang-(1-7) had similar antiangiogenic effects in the chick chorioallantoic membrane, causing a >50% decrease in neovascularization. The Ang-(1-7)-induced reduction in both endothelial cell tubule formation and vessel formation in the chick was completely blocked by the specific Ang-(1-7) receptor antagonist [d-proline(7)]-Ang-(1-7), suggesting that these biological actions are mediated by an AT((1-7)) receptor. Ang-(1-7) significantly reduced vascular endothelial growth factor-A protein and mRNA in tumors from mice treated with the heptapeptide compared with saline controls as well as in the parent A549 human lung cancer cells in culture. These results suggest that Ang-(1-7) may attenuate tumor angiogenesis by reducing vascular endothelial growth factor-A, a primary proangiogenic protein. Taken together, this study shows that Ang-(1-7) exhibits significant antiangiogenic activity and may be a novel therapeutic agent for lung cancer treatment targeting a specific AT((1-7)) receptor.


Angiotensin I/pharmacology , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Peptide Fragments/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , Angiotensin I/administration & dosage , Animals , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/blood supply , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Injections, Subcutaneous , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/drug effects , Peptide Fragments/administration & dosage , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
6.
Cancer Res ; 67(6): 2809-15, 2007 Mar 15.
Article En | MEDLINE | ID: mdl-17363603

Angiotensin-(1-7) [Ang-(1-7)] is an endogenous peptide of the renin-angiotensin system with vasodilator and antiproliferative properties. Our previous studies showed that Ang-(1-7) reduced serum-stimulated growth of human lung cancer cells in vitro through activation of a unique AT((1-7)) receptor. The current study investigates the effect of Ang-(1-7) on lung tumor growth in vivo, using a human lung tumor xenograft model. Athymic mice with tumors resulting from injection of A549 human lung cancer cells were treated for 28 days with either i.v. saline or Ang-(1-7), delivered by implanted osmotic mini-pumps. Treatment with Ang-(1-7) reduced tumor volume by 30% compared with the size before treatment; in contrast, tumor size in the saline-treated animals increased 2.5-fold. These results correlate with a reduction in the proliferation marker Ki67 in the Ang-(1-7)-infused tumors when compared with the saline-infused tumor tissues. Treatment with Ang-(1-7) significantly reduced cyclooxygenase-2 (COX-2) mRNA and protein in tumors of Ang-(1-7)-infused mice when compared with mice treated with saline as well as in the parent A549 human lung cancer cells in tissue culture. These results suggest that Ang-(1-7) may decrease COX-2 activity and proinflammatory prostaglandins to inhibit lung tumor growth. In contrast, the heptapeptide had no effect on COX-1 mRNA in xenograft tumors or A549 cells. Because Ang-(1-7), a peptide with antithrombotic properties, reduces growth through activation of a selective AT((1-7)) receptor, our results suggest that the heptapeptide represents a novel treatment for lung cancer by reducing COX-2.


Adenocarcinoma/drug therapy , Angiotensin I/pharmacology , Cyclooxygenase 2/metabolism , Lung Neoplasms/drug therapy , Peptide Fragments/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Xenograft Model Antitumor Assays
...