Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 151(2): 324-337.e12, 2016 08.
Article in English | MEDLINE | ID: mdl-27178121

ABSTRACT

BACKGROUND & AIMS: High-throughput sequencing technologies have identified thousands of infrequently mutated genes in hepatocellular carcinomas (HCCs). However, high intratumor and intertumor heterogeneity, combined with large numbers of passenger mutations, have made it difficult to identify driver mutations that contribute to the development of HCC. We combined transposon mutagenesis with a high-throughput screen of a small-hairpin RNA (shRNA) library to identify genes and pathways that contribute to HCC development. METHODS: Sleeping beauty transposons were mobilized in livers of transgenic mice predisposed to develop hepatocellular adenoma and HCC owing to expression of the hepatitis B virus surface antigen. This whole-genome mutagenesis technique was used to generate an unbiased catalogue of candidate cancer genes (CCGs). Pooled shRNA libraries targeting 250 selected CCGs then were introduced into immortalized mouse liver cells and the cells were monitored for their tumor-forming ability after injection into nude mice. RESULTS: Transposon-mediated mutagenesis identified 1917 high-confident CCGs and highlighted the importance of Ras signaling in the development of HCC. Subsequent pooled shRNA library screening of 250 selected CCGs validated 27 HCC tumor-suppressor genes. Individual shRNA knockdown of 4 of these genes (Acaa2, Hbs1l, Ralgapa2, and Ubr2) increased the proliferation of multiple human HCC cell lines in culture and accelerated the formation of xenograft tumors in nude mice. The ability of Ralgapa2 to promote HCC cell proliferation and tumor formation required its inhibition of Rala and Ralb. Dual inhibition of Ras signaling via Ral and Raf, using a combination of small-molecule inhibitor RBC8 and sorafenib, reduced the proliferation of HCC cells in culture and completely inhibited their growth as xenograft tumors in nude mice. CONCLUSIONS: In a 2-step forward genetic screen in mice, we identified members of the Ral guanosine triphosphatase-activating protein pathway and other proteins as suppressors of HCC cell proliferation and tumor growth. These proteins might serve as therapeutic targets for liver cancer.


Subject(s)
Carcinoma, Hepatocellular/genetics , GTPase-Activating Proteins/physiology , Genes, Tumor Suppressor , Liver Neoplasms, Experimental/genetics , ral GTP-Binding Proteins/physiology , Animals , Cell Proliferation/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Liver/metabolism , Mice , Mice, Transgenic , Signal Transduction/genetics
2.
PLoS One ; 9(2): e89397, 2014.
Article in English | MEDLINE | ID: mdl-24586749

ABSTRACT

The ecotropic viral integration site 1 (Evi1) oncogenic transcription factor is one of a number of alternative transcripts encoded by the Mds1 and Evi1 complex locus (Mecom). Overexpression of Evi1 has been observed in a number of myeloid disorders and is associated with poor patient survival. It is also amplified and/or overexpressed in many epithelial cancers including nasopharyngeal carcinoma, ovarian carcinoma, ependymomas, and lung and colorectal cancers. Two murine knockout models have also demonstrated Evi1's critical role in the maintenance of hematopoietic stem cell renewal with its absence resulting in the death of mutant embryos due to hematopoietic failure. Here we characterize a novel mouse model (designated Evi1(fl3)) in which Evi1 exon 3, which carries the ATG start, is flanked by loxP sites. Unexpectedly, we found that germline deletion of exon3 produces a hypomorphic allele due to the use of an alternative ATG start site located in exon 4, resulting in a minor Evi1 N-terminal truncation and a block in expression of the Mds1-Evi1 fusion transcript. Evi1(δex3/δex3) mutant embryos showed only a mild non-lethal hematopoietic phenotype and bone marrow failure was only observed in adult Vav-iCre/+, Evi1(fl3/fl3) mice in which exon 3 was specifically deleted in the hematopoietic system. Evi1(δex3/δex3) knockout pups are born in normal numbers but die during the perinatal period from congenital heart defects. Database searches identified 143 genes with similar mutant heart phenotypes as those observed in Evi1(δex3/δex3) mutant pups. Interestingly, 42 of these congenital heart defect genes contain known Evi1-binding sites, and expression of 18 of these genes are also effected by Evi1 siRNA knockdown. These results show a potential functional involvement of Evi1 target genes in heart development and indicate that Evi1 is part of a transcriptional program that regulates cardiac development in addition to the development of blood.


Subject(s)
Alleles , DNA-Binding Proteins/genetics , Genetic Association Studies , Heart Defects, Congenital/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Animals , Animals, Newborn , Base Sequence , Bone Marrow/pathology , DNA-Binding Proteins/chemistry , Disease Models, Animal , Exons , Gene Deletion , Gene Expression Regulation, Developmental , Genes, Lethal , Heart Defects, Congenital/mortality , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Immunophenotyping , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Phenotype , Sequence Alignment , Severity of Illness Index , Transcription Factors/chemistry
3.
Nat Genet ; 46(1): 24-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316982

ABSTRACT

The most common risk factor for developing hepatocellular carcinoma (HCC) is chronic infection with hepatitis B virus (HBV). To better understand the evolutionary forces driving HCC, we performed a near-saturating transposon mutagenesis screen in a mouse HBV model of HCC. This screen identified 21 candidate early stage drivers and a very large number (2,860) of candidate later stage drivers that were enriched for genes that are mutated, deregulated or functioning in signaling pathways important for human HCC, with a striking 1,199 genes being linked to cellular metabolic processes. Our study provides a comprehensive overview of the genetic landscape of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Hepatitis B, Chronic/complications , Liver Neoplasms/genetics , Liver Neoplasms/virology , Mutagenesis , Animals , Carcinoma, Hepatocellular/metabolism , DNA Transposable Elements , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Metabolic Networks and Pathways/genetics , Metabolomics/methods , Mice , Mice, Transgenic , Mutagenesis, Insertional , Pyruvic Acid/metabolism
4.
Proc Natl Acad Sci U S A ; 110(34): E3152-61, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23912186

ABSTRACT

Invasiveness underlies cancer aggressiveness and is a hallmark of malignancy. Most malignant tumors have elevated levels of Tn, an O-GalNAc glycan. Mechanisms underlying Tn up-regulation and its effects remain unclear. Here we show that Golgi-to-endoplasmic reticulum relocation of polypeptide N-acetylgalactosamine-transferases (GalNAc-Ts) drives high Tn levels in cancer cell lines and in 70% of malignant breast tumors. This process stimulates cell adhesion to the extracellular matrix, as well as migration and invasiveness. The GalNAc-Ts lectin domain, mediating high-density glycosylation, is critical for these effects. Interfering with the lectin domain function inhibited carcinoma cell migration in vitro and metastatic potential in mice. We also show that stimulation of cell migration is dependent on Tn-bearing proteins present in lamellipodia of migrating cells. Our findings suggest that relocation of GalNAc-Ts to the endoplasmic reticulum frequently occurs upon cancerous transformation to enhance tumor cell migration and invasiveness through modification of cell surface proteins.


Subject(s)
Acetylgalactosamine/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glycosyltransferases/metabolism , Neoplasm Invasiveness/physiopathology , Neoplasms/physiopathology , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Blotting, Western , Cell Line , Cell Movement/physiology , Cloning, Molecular , Fluorescent Antibody Technique , Glycosylation , Golgi Apparatus/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Neoplasms/metabolism
5.
Proc Natl Acad Sci U S A ; 110(31): E2885-94, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23858473

ABSTRACT

Ecotropic viral integration site-1 (EVI1) is an oncogenic zinc finger transcription factor whose expression is frequently up-regulated in myeloid leukemia and epithelial cancers. To better understand the mechanisms underlying EVI1-associated disease, we sought to define the EVI1 interactome in cancer cells. By using stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics, we could confidently assign 78 proteins as EVI1-interacting partners for FLAG-tagged EVI1. Subsequently, we showed that 22 of 27 tested interacting proteins could coimmunoprecipitate with endogenous EVI1 protein, which represented an 81.5% validation rate. Additionally, by comparing the stable isotope labeling by amino acids in cell culture (SILAC) data with high-throughput yeast two hybrid results, we showed that five of these proteins interacted directly with EVI1. Functional classification of EVI1-interacting proteins revealed associations with cellular transcription machinery; modulators of transcription; components of WNT, TGF-ß, and RAS pathways; and proteins regulating DNA repair, recombination, and mitosis. We also identified EVI1 phosphorylation sites by MS analysis and showed that Ser538 and Ser858 can be phosphorylated and dephosphorylated by two EVI1 interactome proteins, casein kinase II and protein phosphatase-1α. Finally, mutations that impair EVI1 phosphorylation at these sites reduced EVI1 DNA binding through its C-terminal zinc finger domain and induced cancer cell proliferation. Collectively, these combinatorial proteomic approaches demonstrate that EVI1 interacts with large and complex networks of proteins, which integrate signals from various different signaling pathways important for oncogenesis. Comprehensive analysis of the EVI1 interactome has thus provided an important resource for dissecting the molecular mechanisms of EVI1-associated disease.


Subject(s)
DNA-Binding Proteins/metabolism , Mitosis , Neoplasms/metabolism , Oncogene Proteins/metabolism , Recombinational DNA Repair , Transcription Factors/metabolism , Wnt Signaling Pathway , DNA-Binding Proteins/genetics , HeLa Cells , Humans , MDS1 and EVI1 Complex Locus Protein , Neoplasms/genetics , Neoplasms/pathology , Oncogene Proteins/genetics , Phosphorylation/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics
6.
Inflamm Res ; 62(2): 133-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23052185

ABSTRACT

OBJECTIVE AND DESIGN: We investigated the role and regulation of zinc transporters in the activation of the inflammatory response in macrophages. Our exploratory computational study found that Zip14 (SLC39A14) was consistently up-regulated in activated macrophages; we therefore focused subsequently on that gene in the mechanistic study. MATERIAL: The expression and function of Zip14 was assessed in primary macrophages obtained by in-vitro differentiation of monocytes from human blood. METHODS: Primary macrophages were subjected to treatments with lipopolysaccharides, cytokines, chemicals, and pharmacological agents. SLC39A14 and inflammatory cytokine gene expressions were assessed by RT-qPCR. Zip14 siRNA knockdown was performed to explore the gene function. RESULTS: Lipopolysaccharide's inflammatory stimulus was a strong inducer of SLC39A14 mRNA expression in macrophages. This induction was dependent on calcium signaling, GC-rich DNA-binding, and NF-κB down-regulation. Impregnation of lipopolysaccharide-stimulated macrophages with the glucocorticoid dexamethasone further enhanced Zip14 expression while reducing interleukin-6 and tumor necrosis factor-α production. Zip14 knockdown in macrophages attenuated the expression and secretion of cytokines, indicating a buffering function for this zinc transporter. CONCLUSIONS: Collectively, our results identified the zinc transporter Zip14 as expressed downstream of lipopolysaccharide signals in macrophages. Zip14 induction had a regulatory function in cytokine production.


Subject(s)
Cation Transport Proteins/immunology , Macrophages/immunology , Animals , Calcium/metabolism , Cation Transport Proteins/genetics , Cell Differentiation , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Humans , Immunosuppressive Agents/pharmacology , Inflammation/immunology , Interferon-gamma/pharmacology , Lipopolysaccharides , Lung/immunology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , RNA, Messenger/metabolism , Spleen/immunology
7.
Proc Natl Acad Sci U S A ; 109(6): 2168-73, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308434

ABSTRACT

Ecotropic viral integration site 1 (EVI1) is an oncogenic dual domain zinc finger transcription factor that plays an essential role in the regulation of hematopoietic stem cell renewal, and its overexpression in myeloid leukemia and epithelial cancers is associated with poor patient survival. Despite the discovery of EVI1 in 1988 and its emerging role as a dominant oncogene in various types of cancer, few EVI1 target genes are known. This lack of knowledge has precluded a clear understanding of exactly how EVI1 contributes to cancer. Using a combination of ChIP-Seq and microarray studies in human ovarian carcinoma cells, we show that the two zinc finger domains of EVI1 bind to DNA independently and regulate different sets of target genes. Strikingly, an enriched fraction of EVI1 target genes are cancer genes or genes associated with cancer. We also show that more than 25% of EVI1-occupied genes contain linked EVI1 and activator protein (AP)1 DNA binding sites, and this finding provides evidence for a synergistic cooperative interaction between EVI1 and the AP1 family member FOS in the regulation of cell adhesion, proliferation, and colony formation. An increased number of dual EVI1/AP1 target genes are also differentially regulated in late-stage ovarian carcinomas, further confirming the importance of the functional cooperation between EVI1 and FOS. Collectively, our data indicate that EVI1 is a multipurpose transcription factor that synergizes with FOS in invasive tumors.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factors/metabolism , Binding Sites , Cell Adhesion , Chromatin Immunoprecipitation , DNA/genetics , DNA/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , HeLa Cells , Humans , MDS1 and EVI1 Complex Locus Protein , Neoplasm Invasiveness , Protein Binding , Proto-Oncogenes
8.
Cancer Cell ; 19(5): 629-39, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21575863

ABSTRACT

The human gene Ptpn11, which encodes the tyrosine phosphatase Shp2, may act as a proto-oncogene because dominantly activating mutations have been detected in several types of leukemia. Herein we report a tumor-suppressor function of Shp2. Hepatocyte-specific deletion of Shp2 promotes inflammatory signaling through the Stat3 pathway and hepatic inflammation/necrosis, resulting in regenerative hyperplasia and development of tumors in aged mice. Furthermore, Shp2 ablation dramatically enhanced diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) development, which was abolished by concurrent deletion of Shp2 and Stat3 in hepatocytes. Decreased Shp2 expression was detected in a subfraction of human HCC specimens. Thus, in contrast to the leukemogenic effect of dominant-active mutants, Ptpn11/Shp2 has a tumor-suppressor function in liver.


Subject(s)
Adenoma, Liver Cell/enzymology , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Liver/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Tumor Suppressor Proteins/metabolism , Adenoma, Liver Cell/genetics , Adenoma, Liver Cell/pathology , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Cytokines/blood , Cytokines/genetics , Diethylnitrosamine , Gene Expression Regulation , Hepatitis/enzymology , Hepatitis/genetics , Hepatitis/pathology , Humans , Hyperplasia , Inflammation Mediators/blood , Interleukin-6/administration & dosage , Lipopolysaccharides/administration & dosage , Liver/drug effects , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Liver Regeneration , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Mas , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , Time Factors , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
9.
J Biol Chem ; 281(45): 34374-80, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16959766

ABSTRACT

Stat5 and Stat3, two members of the Stat (signal transducer and activator of transcription) family, are known to play critical roles in mammopoiesis/lactogenesis and involution, respectively, in the mammary gland. Phosphotyrosine phosphatase Shp2 has been shown to dephosphorylate and thus inactivate both Stat5 and Stat3 in vitro. Paradoxically, cell culture studies also suggest a positive role of Shp2 in promoting prolactin-stimulated Stat5 activation. We have shown here that selective deletion of Shp2 in mouse mammary glands suppresses Stat5 activity during pregnancy and lactation, resulting in significant impairment of lobulo-alveolar outgrowth and lactation. In contrast, Stat3 activity was slightly up-regulated shortly before/at involution, leading to normal epithelial cell apoptosis/involution in Shp2-deficient mammary gland. Thus, Shp2 acts to promote Stat5 activation by the JAK2.prolactin receptor complex, while negatively modulating Stat3 activity before the onset of involution. This is the first demonstration that Shp2 manipulates Stat5 and Stat3 activities reciprocally in mammary epithelial cells, providing novel insight into the complex mechanisms for regulation of various Stat family members by a cytoplasmic tyrosine phosphatase.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Mammary Glands, Animal/growth & development , Protein Tyrosine Phosphatases/physiology , STAT5 Transcription Factor/metabolism , Animals , Apoptosis , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation , Genotype , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins/genetics , Lactation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Polymerase Chain Reaction , Pregnancy , Prolactin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , Sequence Deletion , Transgenes
10.
Mol Cell Biol ; 26(12): 4664-74, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16738330

ABSTRACT

Liver regeneration is a rapid and concerted response to injury, in which growth factor-generated intracellular signals result in activation of transcription factors, DNA synthesis, and hepatocyte proliferation. However, the link between cytoplasmic signals resulting in proliferative response to liver injury remains to be elucidated. We show here that association of Gab1 adaptor protein and Shp2 tyrosine phosphatase is a critical event at the early phase of liver regeneration. Partial hepatectomy (PH) rapidly and transiently induced assembly of a complex comprising Shp2 and tyrosine-phosphorylated Gab1 in wild-type hepatocytes. Consistently, liver-specific Shp2 knockout (LSKO) and liver-specific Gab1 knockout (LGKO) mice displayed very similar phenotypes of defective liver regeneration triggered by PH, including blunted extracellular signal-regulated kinase 1/2 (Erk1/2) activation, decreased expression of immediate-early genes, and reduced levels of cyclins A, E, and B1, as well as suppression of hepatocyte proliferation. In contrast, the Akt and interleukin-6/Stat3 pathways were up-regulated posthepatectomy in LSKO and LGKO mice, accompanied by improved hepatoprotection. Collectively, this study establishes the physiological significance of the Gab1/Shp2 link in promoting mitogenic signaling through the Erk pathway in mammalian liver regeneration.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Liver Regeneration/physiology , Phosphoproteins/physiology , Protein Tyrosine Phosphatases/physiology , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Cell Proliferation , Cytokines/genetics , DNA/genetics , Down-Regulation , Genes, Immediate-Early , Growth Substances/genetics , Hepatectomy , Hepatocytes/cytology , Hepatocytes/metabolism , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Liver/injuries , Liver/metabolism , Liver Regeneration/genetics , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Phosphoproteins/deficiency , Phosphoproteins/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/deficiency , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism
11.
Nat Med ; 11(5): 567-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15821749

ABSTRACT

Insulin receptor substrate-1 (IRS-1) and IRS-2 are known to transduce and amplify signals emanating from the insulin receptor. Here we show that Grb2-associated binder 1 (Gab1), despite its structural similarity to IRS proteins, is a negative modulator of hepatic insulin action. Liver-specific Gab1 knockout (LGKO) mice showed enhanced hepatic insulin sensitivity with reduced glycemia and improved glucose tolerance. In LGKO liver, basal and insulin-stimulated tyrosine phosphorylation of IRS-1 and IRS-2 was elevated, accompanied by enhanced Akt/PKB activation. Conversely, Erk activation by insulin was suppressed in LGKO liver, leading to defective IRS-1 Ser612 phosphorylation. Thus, Gab1 acts to attenuate, through promotion of the Erk pathway, insulin-elicited signals flowing through IRS and Akt proteins, which represents a novel balancing mechanism for control of insulin signal strength in the liver.


Subject(s)
Insulin/metabolism , Liver/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Blood Chemical Analysis , Blood Glucose , DNA Primers , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Genetic Engineering , Glucose Tolerance Test , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Phosphoproteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...