Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Cell Biol ; 147(5): 921-8, 1999 Nov 29.
Article in English | MEDLINE | ID: mdl-10579713

ABSTRACT

Animal cells divide into two daughter cells by the formation of an actomyosin-based contractile ring through a process called cytokinesis. Although many of the structural elements of cytokinesis have been identified, little is known about the signaling pathways and molecular mechanisms underlying this process. Here we show that the human ECT2 is involved in the regulation of cytokinesis. ECT2 catalyzes guanine nucleotide exchange on the small GTPases, RhoA, Rac1, and Cdc42. ECT2 is phosphorylated during G2 and M phases, and phosphorylation is required for its exchange activity. Unlike other known guanine nucleotide exchange factors for Rho GTPases, ECT2 exhibits nuclear localization in interphase, spreads throughout the cytoplasm in prometaphase, and is condensed in the midbody during cytokinesis. Expression of an ECT2 derivative, containing the NH(2)-terminal domain required for the midbody localization but lacking the COOH-terminal catalytic domain, strongly inhibits cytokinesis. Moreover, microinjection of affinity-purified anti-ECT2 antibody into interphase cells also inhibits cytokinesis. These results suggest that ECT2 is an important link between the cell cycle machinery and Rho signaling pathways involved in the regulation of cell division.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/enzymology , G2 Phase/physiology , Guanine Nucleotide Exchange Factors/metabolism , Mitosis/physiology , Proto-Oncogene Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Antibodies/pharmacology , Antibody Specificity , Cell Division/immunology , Epithelial Cells/metabolism , Growth Inhibitors/biosynthesis , Humans , Microinjections , Protein Structure, Tertiary , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/immunology , Spindle Apparatus/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
2.
Oncogene ; 25(6): 827-37, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16247472

ABSTRACT

The epithelial cell transforming gene 2 (ECT2) protooncogene encodes a Rho exchange factor, and regulates cytokinesis. ECT2 is phosphorylated in G2/M phases, but its role in the biological function is not known. Here we show that two mitotic kinases, Cdk1 and polo-like kinase 1 (Plk1), phosphorylate ECT2 in vitro. We identified an in vitro Cdk1 phosphorylation site (T412) in ECT2, which comprises a consensus phosphospecific-binding module for the Plk1 polo-box domain (PBD). Endogenous ECT2 in mitotic cells strongly associated with Plk1 PBD, and this binding was inhibited by phosphatase treatment. A phosphorylation-deficient mutant form of ECT2, T412A, did not exhibit strong association with Plk1 PBD compared with wild-type (WT) ECT2. Moreover, ECT2 T412A, but not phosphomimic T412D, displayed a diminished accumulation of GTP-bound RhoA compared with WT ECT2, suggesting that phosphorylation of Thr-412 is critical for the catalytic activity of ECT2. Moreover, while overexpression of WT ECT2 or the T412D mutant caused cortical hyperactivity in U2OS cells during cell division, this activity was not observed in cells expressing ECT2 T412A. These results suggest that ECT2 is regulated by Cdk1 and Plk1 in concert.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Division/physiology , G2 Phase/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Binding Sites , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , HeLa Cells , Humans , Mitosis , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Threonine/genetics , Threonine/metabolism , rhoA GTP-Binding Protein/genetics , Polo-Like Kinase 1
3.
Cancer Res ; 52(23): 6501-6, 1992 Dec 01.
Article in English | MEDLINE | ID: mdl-1423296

ABSTRACT

To elucidate the mechanism of the synergistic cytotoxicity of 5-fluorouracil (5-FU) and cis-diamminedichloroplatinum(II) (CDDP), we studied the interaction of these agents using a human squamous carcinoma cell line (HST-1). Exposure to 5-FU for 24 h and to CDDP for 1 h produced a 50% inhibitory concentration of 1.0 micrograms/ml (7.7 microM) and 2.5 micrograms/ml (8.3 microM), respectively. The cytotoxic action of CDDP was augmented, and a greater than additive effect was observed when the cells were exposed to 5-FU (1.0 micrograms/ml; 7.7 microM) for 24 h before the CDDP treatment. This synergistic activity was maximal when the interval between 5-FU and CDDP exceeded 24 h. In contrast, the cytotoxicity of CDDP was attenuated when it preceded the exposure to 5-FU. Thymidine did not alter the 5-FU-CDDP interaction. Evaluation of the kinetics of the removal of DNA interstrand cross-links, measured by alkaline elution, showed a significant reduction of this removal in the cells exposed to 5-FU followed by CDDP with a drug-free interval of 48 h, as compared with cells exposed to CDDP alone, or to 5-FU immediately followed by CDDP, although no differences were found in the formation of DNA interstrand cross-links by CDDP among these cells. No significant differences in the accumulation of intracellular platinum were detected by atomic absorption spectrophotometry. These findings suggest that 5-FU modulates the repair of platinum-DNA adducts, thereby potentiating the antitumor activity of CDDP.


Subject(s)
Cisplatin/metabolism , DNA, Neoplasm/metabolism , Fluorouracil/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Count/drug effects , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA, Neoplasm/drug effects , Drug Administration Schedule , Drug Synergism , Fluorouracil/administration & dosage , Humans , Time Factors , Tumor Cells, Cultured
4.
DNA Cell Biol ; 20(7): 383-90, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11506702

ABSTRACT

The ost protooncogene encodes a guanine nucleotide exchange factor for the Rho family of small GTPases, RhoA and Cdc42. The N-terminal domain of Ost (Ost-N) appears to negatively regulate the oncogenic activity of the protein, as deletion of this domain drastically increases its transforming activity in NIH 3T3 cells. Using a yeast two-hybrid system, we identified five genes encoding proteins that can interact with Ost-N. One of them, designated OSTIP2 (Ost interacting protein 2), encoded a previously uncharacterized protein. The OSTIP2 product is highly expressed in skeletal muscle as a 1.2-kb transcript. Full-length OSTIP2 cDNA contained an ORF of 193 amino acids. Transcription-coupled translation of OSTIP2 cDNA in reticulocyte lysates revealed a protein product of 20 kDa, which corresponded to the predicted size of the protein. Bacterially expressed glutathione S-transferase (GST)-Ostip2 fusion protein efficiently associated in vitro with baculovirus-expressed Ost. Interestingly, expression of Ostip2 in NIH 3T3 cells efficiently induced foci of morphologically transformed cells. Moreover, inoculation of athymic (nude) mice with OSTIP2 transfectants strongly induced tumor formation. These results suggest that Ostip2 is a novel oncoprotein that can interact with the Rho exchange factor Ost.


Subject(s)
Cell Transformation, Neoplastic , Guanine Nucleotide Exchange Factors/metabolism , Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Complementary , HeLa Cells , Humans , Mice , Mice, Nude , Molecular Sequence Data , Oncogene Proteins/metabolism , Saccharomyces cerevisiae , Two-Hybrid System Techniques , rhoA GTP-Binding Protein/metabolism
5.
Oncol Res ; 9(4): 167-72, 1997.
Article in English | MEDLINE | ID: mdl-9268987

ABSTRACT

Pretreatment of 5-fluorouracil (5-FU), but not posttreatment, has been shown to augment the cytotoxicity of cisplatin (CDDP) or even circumvent CDDP resistance by inhibiting repair of platinum-DNA interstrand crosslinks as well as by reducing the cellular glutathione (GSH) contents in CDDP-resistant HST-1/CP0.2 human squamous carcinoma cells. Because exogenous thymidine, which compensates for 5-FU-mediated inhibition of de novo DNA synthesis via salvage pathway, did not affect this schedule-dependent synergism, the modulatory effect of 5-FU on CDDP resistance would be attributed to the 5-FU-induced RNA damage. We therefore examined the effect of 5-FU on the steady-state levels of messenger RNA (mRNA) of a human excision repair gene ERCC1 and gamma-glutamylcysteine synthetase (gamma-GCS) gene coding for a rate-limiting enzyme for GSH synthesis. The HST-1/ CP0.2 cells were found to have significantly more mRNA expression of these respective genes than do parental HST-1 cells. In these cells, 5-FU pretreatment progressively inhibited mRNA expression of both ERCC1 and gamma-GCS after removal of 5-FU, without affecting glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA. A maximal mRNA suppression was observed at 48 h posttreatment. Such 5-FU-induced suppression of mRNA transcripts of these genes seems to be consistent with its inhibitory activity on DNA repair capacity and cellular GSH contents. In contrast, 5-FU did not reduce the level of glutathione-S-transferase-pi (GST-pi) or DNA topoisomerase 1 mRNA. Although not convinced, our data suggest that 5-FU, when incorporated into RNA, may inhibit both GSH synthesis and repair of platinum-DNA adducts by downregulating the ERCC1 and gamma-GCS genes, thereby enhancing antitumor activity of CDDP and reversing resistance to CDDP in HST-1/CP0.2 cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins , Endonucleases , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glutamate-Cysteine Ligase/genetics , Proteins/genetics , Cisplatin/pharmacology , DNA Repair/drug effects , DNA Topoisomerases, Type I/genetics , Drug Resistance, Neoplasm , Humans , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Thymidine/pharmacology
6.
Anticancer Res ; 15(2): 405-9, 1995.
Article in English | MEDLINE | ID: mdl-7763013

ABSTRACT

BACKGROUND: We studied the modulatory effects of 7-ethyl-10-hydroxy-camptothecin (SN-38), an active metabolite of 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin, on the antitumor activities of clinically important anticancer drugs including cisplatin (CDDP), 5-fluorouracil (5-FU), mitomycin C (MMC), etoposide (VP-16), and adriamycin (ADR). MATERIALS AND METHODS: The HST-1 human carcinoma cells were treated with graded concentrations of these anticancer drugs either alone or in combination with IC50 concentration of SN-38, administered in several different treatment schedules, and antitumor activity was evaluated by the growth inhibition assay. RESULTS: SN-38 potentiated the antitumor activity of CDDP in all schedules with a maximal effect observed with a simultaneous administration, while SN-38 enhanced the cytotoxicity of MMC, 5-FU, or VP-16 only in certain schedules. By contrast, SN-38 attenuated the anticancer effect of ADR in all schedules. CONCLUSIONS: These results demonstrate that SN-38 may have the advantage of augmenting the anticancer activity in combination with CDDP, MMC, 5-FU, and VP-16, depending on the schedule of administration, and should thus be incorporated into the design of a clinical trial for obtaining maximal therapeutic synergy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Topoisomerase I Inhibitors , Camptothecin/pharmacology , Carcinoma, Squamous Cell/metabolism , Cisplatin/pharmacology , Doxorubicin/pharmacology , Etoposide/pharmacology , Fluorouracil/pharmacology , Humans , Infant, Newborn , Irinotecan , Mitomycin/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Stem Cell Assay
7.
Fukuoka Igaku Zasshi ; 83(1): 43-50, 1992 Jan.
Article in Japanese | MEDLINE | ID: mdl-1547989

ABSTRACT

A 34-year-old woman had developed frequent episodes of disorientation at night since a year ago. Her blood ammonia level was found to be markedly increased. Serum amino acid pattern and biochemical analysis of urea cycle enzymes in the liver indicated type II citrullinemia. Pancreatolithiasis was found by ultrasonography and CT scan. After a short remission following treatment by protein restriction and oral administration of sodium benzoate, she had disturbed consciousness due to exaggerated hyperammonemia and died of severe brain edema. In this report, efficacy of the treatment for hyperammonemia and histopathological findings are discussed.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Citrulline/blood , Pancreatitis/complications , Adult , Chronic Disease , Female , Humans
9.
Int J Cancer ; 61(2): 206-13, 1995 Apr 10.
Article in English | MEDLINE | ID: mdl-7705949

ABSTRACT

The roles of activated ras and src oncogene products in the acquisition of fully neoplastic phenotype by human gallbladder adenocarcinoma cells were investigated by co-transfecting non-tumorigenic HAG-I human gallbladder carcinoma cells with the pSV2neo plasmid and a plasmid carrying either activated c-H-ras or v-src oncogene. G418-resistant clones were isolated and assessed for the acquisition of anchorage-independent growth potential. Neither the 10 established clones transfected with pSV2neo alone nor the 17 clones transfected with activated c-H-ras, including 4 clones expressing the mutated p21H-ras protein, could form colonies in soft agar. By contrast, out of 10 clones transfected with v-src, 2 formed colonies in soft agar and produced tumors in athymic nude mice, the resulting progressive neoplasms being poorly differentiated adenocarcinomas. These tumorigenic clones were shown to have v-src DNA and mRNA levels with p60v-src protein, but there were no significant chromosomal alterations following tumorigenic conversion. Moreover, herbimycin A, a selective src-kinase inhibitor, markedly reduced clonogenic growth of these cells in soft agar rather than monolayer growth, suggesting that anchorage-independent growth of the v-src-transformed HAG-I cells might be driven directly by p60v-src kinase activity. Taken together, our data suggest that the fully neoplastic conversion of HAG-I cells depends on src-related tyrosine-kinase activity, but not solely on the function mediated by activated ras, thus providing evidence of an src-related signaling pathway for the acquisition of tumorigenic potential by human gallbladder adenocarcinoma cells.


Subject(s)
Adenocarcinoma/genetics , Cell Transformation, Neoplastic/genetics , Gallbladder Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, ras , Genes, src , Adenocarcinoma/pathology , Animals , Benzoquinones , Blotting, Northern , Blotting, Southern , Cell Adhesion/physiology , Cell Division/drug effects , Cell Division/physiology , Disease Progression , Gallbladder Neoplasms/pathology , Humans , Immunoblotting , Karyotyping , Lactams, Macrocyclic , Mice , Mice, Nude , Phenotype , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinones/pharmacology , Rifabutin/analogs & derivatives , Transfection
10.
Int J Cancer ; 62(1): 70-5, 1995 Jul 04.
Article in English | MEDLINE | ID: mdl-7601570

ABSTRACT

The combination of cis-diamminedichloroplatinum(II) (CDDP) and 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11), a topoisomerase-I inhibitor, has been shown to be synergistic in vitro and clinically active against several human cancers refractory to chemotherapy. To elucidate the mechanism of the synergistic cytotoxicity of CDDP and 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of CPT-11, we studied the interaction of these agents using an HST-1 human squamous-carcinoma cell line. Cells were exposed to the IC50 concentration of SN-38 (5.0 ng/ml) for 1 hr and various concentrations of CDDP for 1 hr in several different treatment schedules. SN-38 augmented the anti-tumor activity of CDDP in all schedules, with maximal synergy observed with simultaneous administration. Evaluation of the kinetics of the removal of DNA interstrand cross-links, measured by alkaline elution, showed significant reduction of this removal in the cells exposed to SN-38 and CDDP, as compared with the cells exposed to CDDP alone. No differences, however, were found in the initially attained level of DNA interstrand cross-links induced by CDDP between these cells. Moreover, the intracellular accumulation of platinum measured by atomic-absorption spectrophotometry, was virtually identical between these cells. These results indicate that SN-38 can modulate the removal of platinum-DNA adducts, thereby potentiating the cytotoxicity of CDDP, suggesting a critical role for topoisomerase I in the repair of DNA interstrand cross-links.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Cisplatin/pharmacology , DNA Repair/drug effects , Camptothecin/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cisplatin/metabolism , DNA Damage , DNA Topoisomerases, Type I/physiology , Humans , Irinotecan , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Tumor Cells, Cultured
11.
Int J Cancer ; 76(3): 423-9, 1998 May 04.
Article in English | MEDLINE | ID: mdl-9579582

ABSTRACT

To investigate whether interferons (IFNs) selectively suppress the growth of solid tumor cells with elevated protein tyrosine kinase (PTK) activity, we evaluated the effect of recombinant IFN-alpha2a and IFN-gamma on the proliferative and neoplastic potentials triggered by p60v-src using v-src-transformed HAG-1 human epithelial cells. When compared with control cells harboring the pSV2neo gene, the monolayer growth of v-src-transformed cell lines was inhibited by both recombinant IFNs, in a dose-dependent manner, whereas growth of ras-transfected cell lines was not affected. Moreover, IFNs markedly reduced the clonogenic growth of v-src-transformed cells in soft-agar rather than monolayer growth, suggesting the preferential activity of IFNs on anchorage-independent growth. Pretreatment of cells with Src or the Src-like PTK inhibitor herbimycin A or radicicol, alleviated dose-dependently the growth-inhibitory activity of IFN-alpha2a against v-src-transformed cells, suggesting that IFNs may share a common inhibitory pathway with Src PTK inhibitors. Accordingly, like herbimycin A, IFNs were found to reduce tyrosine phosphorylation of p60v-src and suppressed in vitro p60v-src kinase activity in v-src-transformed cells. Our data, together with the fact that IFNs inhibit the growth potential driven by Src but not by activated Ras, suggest that inhibition of signal transduction pathway through Src to downstream transduction events may be a primary mechanism of IFN-induced anti-prolifeative and anti-tumoral activity.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Genes, src/genetics , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Protein-Tyrosine Kinases/drug effects , src-Family Kinases/drug effects , Adenocarcinoma/enzymology , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , Epithelial Cells/enzymology , Gallbladder Neoplasms/enzymology , Genes, ras/genetics , Humans , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Transfection , Tyrosine/metabolism , src-Family Kinases/antagonists & inhibitors
12.
J Immunol ; 149(3): 940-8, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1378869

ABSTRACT

We have generated a number of EBV-transformed B cell lines producing human mAb against human T cell leukemia virus type 1 (HTLV-1) from the peripheral blood B lymphocytes obtained from patients with HTLV-1-associated myelopathy/tropical spastic paraparesis. Various synthetic peptides corresponding to antigenic regions of HTLV-1 gag and env proteins were used for the screening of antibodies in ELISA. In our study, four IgG mAb to the gag p19 amino acids 100 to 130, and 5 IgG mAb to the env p46 amino acids 175 to 199 were characterized. An immunofluorescence assay showed that all of these mAb specifically bound to the surface of HTLV-1-bearing cell lines. Among these mAb, one anti-gp46 mAb, designated KE36-11, neutralized the infectivity of HTLV-1 as determined by both the inhibition of HTLV-1-induced syncytium formation and transformation assays in vitro. An antibody-binding assay using overlapping oligopeptides revealed that KE36-11 recognized a new epitope locating between the gp46 amino acid sequence 187-193 (Ala-Pro-Pro-Leu-Leu-Pro-His). Another anti-gp46 mAb, designated KE36-7, showed antibody-dependent cellular cytotoxicity against HTLV-1-bearing cell line. KE36-7 bound strongly to the 10-mer peptide-gp46 187-196, and weakly to peptides containing the gp46 amino acid sequence 191-196 (Leu-Pro-His-Ser-Asn-Leu). These two epitopes, which are associated with HTLV-1 neutralization and antibody-dependent cellular cytotoxicity, are thus the first epitopes identified in human HTLV-1 infection. It is possible that passive immunization of humans with these two human mAb are effective on the protection of HTLV-1 infection in vivo.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Deltaretrovirus Antibodies/immunology , Deltaretrovirus Antigens/immunology , Human T-lymphotropic virus 1/immunology , Amino Acid Sequence , Cell Transformation, Viral , Epitopes , Gene Products, env/immunology , Gene Products, gag/immunology , Herpesvirus 4, Human , Humans , Molecular Sequence Data , Neutralization Tests , Peptides/chemistry , Peptides/immunology
13.
Biol Pharm Bull ; 17(5): 668-71, 1994 May.
Article in English | MEDLINE | ID: mdl-7920430

ABSTRACT

To search for possible anti-tumor-promoters, we carried out a primary screening of 21 cucurbitane triterpenoids using an in vitro assay system. Of these triterpenoids, scandenoside R6 (6), 23,24-dihydrocucurbitacin F (14), 25-acetyl-23,24-dihydrocucurbitacin F (15), 2-O-beta-D-glucopyranosyl-23,24-dihydrocucurbitacin F (17) and cucurbitacin F (18) exhibited significant inhibitory effects on Epstein-Barr virus (EBV) activation induced by the tumor promoter, 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Further, compounds 14 and 17 exhibited remarkable anti-tumor-promotion effects on mouse skin tumor promotion in an in vivo two-stage carcinogenesis test.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Herpesvirus 4, Human/drug effects , Plants, Medicinal/chemistry , Skin Neoplasms/prevention & control , Triterpenes/pharmacology , Virus Activation/drug effects , Animals , Carbohydrate Sequence , Drug Screening Assays, Antitumor , Female , Mice , Mice, Inbred ICR , Molecular Sequence Data , Papilloma/microbiology , Papilloma/prevention & control , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate/pharmacology , Triterpenes/chemistry , Triterpenes/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL