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.
Cell Signal ; 17(4): 461-71, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15601624

ABSTRACT

The Dbl-like guanine nucleotide exchange factors (GEFs) have been implicated in direct activation of the Rho family of small GTPases. We previously isolated transforming immortalized mammary (TIM) as a Dbl-like protein. Here, we show that, when expressed in cells, TIM was a potent activator of RhoA. Like activated Rho proteins, expression of TIM potentiated the serum response factor (SRF)- and AP-1-regualted transcriptional activities and activated the SAPK/JNK signaling pathway. In NIH 3T3 cells, TIM induced transforming foci, which was inhibited by the ROCK inhibitor Y-27632 or the dominant negative mutants of Rho proteins. Expression of TIM led to pronounced changes in cell shape and organization of the actin cytoskeleton, including the formation of thick stress fibers at the cell periphery and cell rounding. TIM also promoted redistribution of vinculin-enriched focal adhesions at the cell periphery and increased the phosphorylation of myosin light chain (MLC). These results, taken together, suggest that TIM acts as an upstream regulator for the RhoA/ROCK-mediated cellular functions.


Subject(s)
Cytoskeleton/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Proto-Oncogene Proteins/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Cell Shape , Cytoskeleton/pathology , Humans , Intracellular Signaling Peptides and Proteins , Mice , NIH 3T3 Cells , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rho Guanine Nucleotide Exchange Factors , Serum Response Factor/metabolism , Signal Transduction , rho-Associated Kinases , rhoA GTP-Binding Protein/genetics
2.
J Biol Chem ; 279(8): 7169-79, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14645260

ABSTRACT

The human ECT2 protooncogene encodes a guanine nucleotide exchange factor for the Rho GTPases and regulates cytokinesis. Although the oncogenic form of ECT2 contains an N-terminal truncation, it is not clear how the structural abnormality of ECT2 causes malignant transformation. Here we show that both the removal of the negative regulatory domain and alteration of subcellular localization are required to induce the oncogenic activity of ECT2. The transforming activity of oncogenic ECT2 was strongly inhibited by dominant negative Rho GTPases, suggesting the involvement of Rho GTPases in ECT2 transformation. Although deletion of the N-terminal cell cycle regulator-related domain (N) of ECT2 did not activate its transforming activity, removal of the small central domain (S), which contains two nuclear localization signals (NLSs), significantly induced the activity. The ECT2 N domain interacted with the catalytic domain and significantly inhibited the focus formation by oncogenic ECT2. Interestingly, the introduction of the NLS mutations in the S domain of N-terminally truncated ECT2 dramatically induced the transforming activity of this otherwise non-oncogenic derivative. Among the known Rho GTPases expressed in NIH 3T3 cells, RhoA was predominantly activated by oncogenic ECT2 in vivo. Therefore, the mislocalization of structurally altered ECT2 might cause the untimely activation of cytoplasmic Rho GTPases leading to the malignant transformation.


Subject(s)
Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/physiology , rho GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , COS Cells , Catalytic Domain , Cell Transformation, Neoplastic , Cytoplasm/metabolism , Cytoskeleton/metabolism , DNA, Complementary/metabolism , Gene Deletion , Genes, Reporter , Genetic Vectors , Luciferases/metabolism , Mice , Microscopy, Video , Models, Biological , Mutation , NIH 3T3 Cells , Protein Structure, Tertiary , Signal Transduction , Time Factors , Transcriptional Activation , Transfection
3.
J Cell Biochem ; 90(5): 892-900, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14624449

ABSTRACT

The ECT2 protooncogene encodes a guanine nucleotide exchange factor for the Rho family of small GTPases. ECT2 contains motifs of cell cycle regulators at its N-terminal domain. We previously showed that ECT2 plays a critical role in cytokinesis. Here, we report a potential role of XECT2, the Xenopus homologue of the human ECT2, in spindle assembly in cell-free Xenopus egg extracts. Cloned XECT2 cDNA encodes a 100 kDa protein closely related to human ECT2. XECT2 is specifically phosphorylated in M phase extracts. Affinity-purified anti-XECT2 antibody strongly inhibited mitosis in Xenopus cell-free extracts. Instead of bipolar spindles, where chromosomes are aligned at the metaphase plane in control extracts, the addition of anti-XECT2 resulted in the appearance of abnormal spindles including monopolar and multipolar spindles as well as bipolar spindles with misaligned chromosomes. In these in vitro synthesized spindle structures, XECT2 was found to tightly associate with mitotic spindles. The N-terminal half of XECT2 lacking the catalytic domain also strongly inhibited spindle assembly in vitro, resulting in the formation of mitotic spindles with a low density. Among the representative Rho GTPases, a dominant-negative form of Cdc42 strongly inhibited spindle assembly in vitro. These results suggest that the Rho family GTPase Cdc42 and its exchange factor XECT2 are critical regulators of spindle assembly in Xenopus egg extracts.


Subject(s)
Mitosis/physiology , Ovum/physiology , Proto-Oncogene Proteins/physiology , Spindle Apparatus/metabolism , Xenopus laevis/physiology , cdc42 GTP-Binding Protein/physiology , Amino Acid Sequence , Animals , Cell Extracts , Chromosomes/genetics , Female , Male , Molecular Sequence Data , Ovum/cytology , Phosphorylation , Sequence Homology, Amino Acid , Spermatozoa/cytology
4.
J Cell Biochem ; 90(4): 819-36, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14587037

ABSTRACT

The ECT2 protooncogene plays a critical role in cytokinesis, and its C-terminal half encodes a Dbl homology-pleckstrin homology module, which catalyzes guanine nucleotide exchange on the Rho family of small GTPases. The N-terminal half of ECT2 (ECT2-N) contains domains related to the cell cycle regulator/checkpoint control proteins including human XRCC1, budding yeast CLB6, and fission yeast Cut5. The Cut5-related domain consists of two BRCT repeats, which are widespread to repair/checkpoint control proteins. ECT2 is ubiquitously expressed in various tissues and cell lines, but elevated levels of ECT2 expression were found in various tumor cell lines and rapidly developing tissues in mouse embryos. Consistent with these findings, induction of ECT2 expression was observed upon stimulation by serum or various growth factors. In contrast to other oncogenes whose expression is induced early in G1, ECT2 expression was induced later, coinciding with the initiation of DNA synthesis. To test the role of the cell cycle regulator/checkpoint control protein-related domains of ECT2 in cytokinesis, we expressed various ECT2 derivatives in U2OS cells, and analyzed their DNA content by flow cytometry. Expression of the N-terminal half of ECT2, which lacks the catalytic domain, generated cells with more than 4N DNA content, suggesting that cytokinesis was inhibited in these cells. Interestingly, ECT2-N lacking the nuclear localization signals inhibited cytokinesis more strongly than the derivatives containing these signals. Mutational analyses revealed that the XRCC1, CLB6, and BRCT domains in ECT2-N are all essential for the cytokinesis inhibition by ECT2-N. These results suggest that the XRCC1, CLB6, and BRCT domains of ECT2 play a critical role in regulating cytokinesis.


Subject(s)
Growth Substances/pharmacology , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Blood Proteins/pharmacology , Cell Division/drug effects , Cell Line , Cloning, Molecular , DNA/analysis , DNA/genetics , DNA Repair , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice , Mitosis/drug effects , Molecular Sequence Data , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , S Phase/drug effects , Sequence Alignment
5.
Cell Biol Int ; 27(5): 415-21, 2003.
Article in English | MEDLINE | ID: mdl-12758089

ABSTRACT

Detachment of anchorage-dependent normal epithelial cells from their substratum causes the type of apoptosis known as anoikis, whereas malignant cells can proliferate independently of anchorage. Because src and ras oncogenes are activated in many human cancers, we investigated their role and downstream signaling pathways in anoikis resistance, using HAG-1 human epithelial cells transfected with v-src or activated H-ras. Consequently, anchorage-dependent mock- or ras-transfected cells underwent anoikis. In contrast, anchorage-independent v-Src-transformed cells did not exhibit such apoptotic features. Focal adhesion kinase (FAK), a transducer of integrin, was only activated in v-Src-transformed cells. Herbimycin A, an Src kinase inhibitor, reduced tyrosyl phosphorylation of FAK and reversed resistance to anoikis. However, both protein kinase C (PKC) and phophatidylinositol-3 (PI-3) kinase inhibitors failed to induce anoikis. These data suggest that the ability of activated Src to prevent anoikis may be mediated by Src to a downstream signaling pathway involving FAK, but not Ras, PI-3 kinase, or PKC.


Subject(s)
Anoikis/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gallbladder/metabolism , Gene Expression Regulation , Genes, ras , Genes, src/physiology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Gallbladder/drug effects , Gene Expression Regulation/physiology , Genes, ras/genetics , Genes, ras/physiology , Genes, src/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...