Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 71(12): 4260-8, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21512136

ABSTRACT

LIN28B is a homologue of LIN28 that induces pluripotency when expressed in conjunction with OCT4, SOX2, and KLF4 in somatic fibroblasts. LIN28B represses biogenesis of let-7 microRNAs and is implicated in both development and tumorigenesis. Recently, we have determined that LIN28B overexpression occurs in colon tumors. We conducted a comprehensive analysis of LIN28B protein expression in human colon adenocarcinomas. We found that LIN28B overexpression correlates with reduced patient survival and increased probability of tumor recurrence. To elucidate tumorigenic functions of LIN28B, we constitutively expressed LIN28B in colon cancer cells and evaluated tumor formation in vivo. Tumors with constitutive LIN28B expression exhibit increased expression of colonic stem cell markers LGR5 and PROM1, mucinous differentiation, and metastasis. Together, our findings point to a function for LIN28B in promoting colon tumor pathogenesis, especially metastasis.


Subject(s)
Colonic Neoplasms/etiology , RNA-Binding Proteins/physiology , Adult , Aged , Aged, 80 and over , Animals , Cell Differentiation , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Disease Progression , Female , Gene Expression Profiling , Humans , Kruppel-Like Factor 4 , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplastic Stem Cells/physiology
2.
Cancer Res ; 71(6): 2172-82, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21252116

ABSTRACT

Insulin-like growth factor 2 mRNA-binding protein-1 (IMP-1) is an oncofetal protein that binds directly to and stabilizes oncogenic c-Myc and regulates, in turn, its posttranscriptional expression and translation. In contrast to normal adult tissue, IMP-1 is reexpressed and/or overexpressed in human cancers. We show that knockdown of c-Myc in human colon cancer cell lines increases the expression of mature let-7 miRNA family members and downregulates several of its mRNA targets: IMP-1, Cdc34, and K-Ras. We further show that loss of IMP-1 inhibits Cdc34, Lin-28B, and K-Ras, suppresses SW-480 cell proliferation and anchorage-independent growth, and promotes caspase- and lamin-mediated cell death. We also found that IMP-1 binds to the coding region and 3'UTR of K-Ras mRNA. RNA microarray profiling and validation by reverse transcription PCR reveals that the p53-inducible proapoptotic protein CYFIP2 is upregulated in IMP-1 knockdown SW480 cells, a novel finding. We also show that overexpression of IMP-1 increases c-Myc and K-Ras expression and LIM2405 cell proliferation. Furthermore, we show that loss of IMP-1 induces Caspase-3- and PARP-mediated apoptosis, and inhibits K-Ras expression in SW480 cells, which is rescued by CYFIP2 knockdown. Importantly, analysis of 228 patients with colon cancers reveals that IMP-1 is significantly upregulated in differentiated colon tumors (P ≤ 0.0001) and correlates with K-Ras expression (r = 0.35, P ≤ 0.0001) relative to adjacent normal mucosa. These findings indicate that IMP-1, interrelated with c-Myc, acts upstream of K-Ras to promote survival through a novel mechanism that may be important in colon cancer pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colonic Neoplasms/metabolism , RNA-Binding Proteins/metabolism , ras Proteins/metabolism , 3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Cell Survival , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Humans , Immunoblotting , Male , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Up-Regulation , ras Proteins/genetics
4.
Cancer Cell ; 5(4): 375-87, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15093544

ABSTRACT

Activating mutations in the ras oncogene are not considered sufficient to induce abnormal cellular proliferation in the absence of cooperating oncogenes. We demonstrate that the conditional expression of an endogenous K-ras(G12D) allele in murine embryonic fibroblasts causes enhanced proliferation and partial transformation in the absence of further genetic abnormalities. Interestingly, K-ras(G12D)-expressing fibroblasts demonstrate attenuation and altered regulation of canonical Ras effector signaling pathways. Widespread expression of endogenous K-ras(G12D) is not tolerated during embryonic development, and directed expression in the lung and GI tract induces preneoplastic epithelial hyperplasias. Our results suggest that endogenous oncogenic ras is sufficient to initiate transformation by stimulating proliferation, while further genetic lesions may be necessary for progression to frank malignancy.


Subject(s)
Cell Transformation, Neoplastic , Congenital Abnormalities/genetics , Fibroblasts/pathology , Gene Expression Regulation, Developmental/physiology , Genes, ras/physiology , Neoplasms/genetics , Animals , Cell Cycle , Cell Division , Cellular Senescence , Congenital Abnormalities/pathology , Crosses, Genetic , Cyclin-Dependent Kinase Inhibitor p16 , Embryo, Mammalian/cytology , Female , Fibroblasts/metabolism , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neoplasms/pathology , Stem Cells/pathology , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Viral Proteins/metabolism
5.
Cancer Cell ; 4(6): 437-50, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14706336

ABSTRACT

To evaluate the role of oncogenic RAS mutations in pancreatic tumorigenesis, we directed endogenous expression of KRAS(G12D) to progenitor cells of the mouse pancreas. We find that physiological levels of Kras(G12D) induce ductal lesions that recapitulate the full spectrum of human pancreatic intraepithelial neoplasias (PanINs), putative precursors to invasive pancreatic cancer. The PanINs are highly proliferative, show evidence of histological progression, and activate signaling pathways normally quiescent in ductal epithelium, suggesting potential therapeutic and chemopreventive targets for the cognate human condition. At low frequency, these lesions also progress spontaneously to invasive and metastatic adenocarcinomas, establishing PanINs as definitive precursors to the invasive disease. Finally, mice with PanINs have an identifiable serum proteomic signature, suggesting a means of detecting the preinvasive state in patients.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genes, ras/physiology , Mutation , Pancreatic Neoplasms/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Carcinoma, Pancreatic Ductal/metabolism , Cyclooxygenase 2 , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Isoenzymes/metabolism , Matrix Metalloproteinase 7/metabolism , Membrane Proteins , Mice , Neoplasm Metastasis , Neoplasm Staging , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Transcription Factor HES-1
SELECTION OF CITATIONS
SEARCH DETAIL
...