Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Oncogene ; 32(1): 27-38, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-22349823

ABSTRACT

The epidermal growth factor receptor (EGFR) has an essential role in multiple signaling pathways, including cell proliferation and migration, through extracellular ligand binding and subsequent activation of its intracellular tyrosine kinase (TK) domain. The non-small cell lung cancer (NSCLC)-associated EGFR mutants, L858R and G719S, are constitutively active and oncogenic. They display sensitivity to TK inhibitors, including gefitinib and erlotinib. In contrast, the secondary mutation of the gatekeeper residue, T790M, reportedly confers inhibitor resistance on the oncogenic EGFR mutants. In this study, our biochemical analyses revealed that the introduction of the T790M mutation confers gefitinib resistance on the G719S mutant. The G719S/T790M double mutant has enhanced activity and retains high gefitinib-binding affinity. The T790M mutation increases the ATP affinity of the G719S mutant, explaining the acquired drug resistance of the double mutant. Structural analyses of the G719S/T790M double mutant, as well as the wild type and the G719S and L858R mutants, revealed that the T790M mutation stabilizes the hydrophobic spine of the active EGFR-TK conformation. The Met790 side chain of the G719S/T790M double mutant, in the apo form and gefitinib- and AMPPNP-bound forms, adopts different conformations that explain the accommodation of these ligands. In the L858R mutant structure, the active-site cleft is expanded by the repositioning of Phe723 within the P-loop. Notably, the introduction of the F723A mutation greatly enhanced the gefitinib sensitivity of the wild-type EGFR in vivo, supporting our hypothesis that the expansion of the active-site cleft results in enhanced gefitinib sensitivity. Taken together, our results provide a structural basis for the altered drug sensitivities caused by distinct NSCLC-associated EGFR mutations.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Mutation , Quinazolines/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Drug Screening Assays, Antitumor , ErbB Receptors/chemistry , Gefitinib , Humans , Lung Neoplasms/genetics , Protein Conformation , Protein-Tyrosine Kinases/genetics
2.
Oncogene ; 26(55): 7620-267, 2007 Dec 06.
Article in English | MEDLINE | ID: mdl-17599059

ABSTRACT

The tumor suppressor adenomatous polyposis coli (APC) is mutated in sporadic and familial colorectal tumors. APC interacts with the Rac1-specific guanine-nucleotide exchange factor (GEF) Asef, which contains an APC-binding region (ABR) in addition to Dbl homology (DH), Pleckstrin (PH) and Src homology 3 (SH3) domains. APC stimulates the GEF activity of Asef, and thereby regulates cell adhesion and migration. Here, we have identified a second Asef, termed Asef2, that shows significant structural and functional similarities to Asef. We found that both the N-terminal ABR and SH3 domains of Asef2 are responsible for its interaction with APC. When expressed in HeLa cells, a mutant Asef2 lacking the ABR and SH3 domains, Asef2-DeltaABR/SH3, induced increases in the levels of the active forms of Rac1 and Cdc42. Full-length Asef2 also showed this activity when co-transfected with truncated mutant APC expressed in colorectal tumor cells. Consistent with this, either Asef2-DeltaABR/SH3 or Asef2 plus truncated mutant APC stimulated lamellipodia formation in MDCK cells and filopodia formation in HeLa cells. Furthermore, RNA interference experiments showed that Asef2 is required for migration of colorectal tumor cells expressing truncated APC. These results suggest that similar to Asef, Asef2 plays an important role in cell migration, and that Asef2 activated by truncated mutant APC is required for aberrant migration of colorectal tumor cells.


Subject(s)
Cell Movement , Colorectal Neoplasms/pathology , Guanine Nucleotide Exchange Factors/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Amino Acid Sequence , Animals , Cell Movement/genetics , Chlorocebus aethiops , Colorectal Neoplasms/metabolism , Dogs , Guanine Nucleotide Exchange Factors/analysis , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Molecular Sequence Data , Mutation , Protein Interaction Mapping , Pseudopodia/physiology , Rho Guanine Nucleotide Exchange Factors , src Homology Domains/genetics
3.
Oncogene ; 23(29): 5023-31, 2004 Jun 24.
Article in English | MEDLINE | ID: mdl-15064721

ABSTRACT

Cellular transformation occurs only in cells that express both ErbB1 and ErbB4 receptors, but not in cells expressing only one or the other of these receptors. However, when both receptors are coexpressed and ligand-stimulated, they interact with virtually the same adaptor/effector proteins as when expressed singly. To reveal the underlying regulatory mechanism of the kinase/phosphatase network in ErbB homo- and heterodimer receptor signaling, extracellular signal-regulated kinase (ERK) and Akt activities were evaluated in the presence of several enzyme inhibitors in ligand-induced cells expressing ErbB1 (E1), ErbB4 (E4), and ErbB1/ErbB4 (E1/4) receptor. The PP2A inhibitor okadaic acid showed receptor-specific inhibitory profiles for ERK and Akt activities. Moreover, B-Raf isolated only from E1/4 cells could induce in vitro phosphorylation for MEK; this B-Raf kinase activity was abolished by pretreatment of the cells with okadaic acid. Our study further showed that the E1/4 cell-specific B-Raf activity was stimulated by PLC gamma and subsequent Rap1 activation. The present study suggests that B-Raf kinase, which was specifically activated in the cells coexpressing ErbB1 and ErbB4 receptors, elevates total ERK activity within the cell and, therefore, can induce cellular transformation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , ErbB Receptors/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-raf/metabolism , Animals , CHO Cells , Cricetinae , Enzyme Activation , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neuregulin-1/pharmacology , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, ErbB-4 , Signal Transduction
4.
Mol Genet Genomics ; 271(3): 317-24, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14966659

ABSTRACT

Resistance to streptomycin in bacterial cells often results from a mutation in the rpsL gene that encodes the ribosomal protein S12. We found that a particular rpsL mutation (K87E), newly identified in Escherichia coli, causes aberrant protein synthesis activity late in the growth phase. While protein synthesis decreased with age in cells in the wild-type strain, it was sustained at a high level in the mutant, as determined using living cells. This was confirmed using an in vitro protein synthesis system with poly(U) and natural mRNAs (GFP mRNA and CAT mRNA). Other classical rpsL mutations (K42N and K42T) tested did not show such an effect, indicating that this novel characteristic is typical of ribosomes bearing the K87E mutant form of S12, although the K87E mutation conferred the streptomycin resistance and error-restrictive phenotypes also seen with the K42N and K42T mutations. The K87E (but not K42N or K42T) mutant ribosomes exhibited increased stability of the 70S complex in the presence of low concentrations of magnesium. We propose that the aberrant activation of protein synthesis at the late growth phase is caused by the increased stability of the ribosome.


Subject(s)
Chloramphenicol O-Acetyltransferase/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Luminescent Proteins/metabolism , Mutation/genetics , Ribosomal Proteins/genetics , Chloramphenicol O-Acetyltransferase/genetics , Drug Resistance, Microbial , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins , Gene Expression Regulation, Bacterial/drug effects , Green Fluorescent Proteins , Luminescent Proteins/genetics , Magnesium/pharmacology , Molecular Sequence Data , Mutagenesis, Site-Directed , Poly U , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Protein S9 , Ribosomal Proteins/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Streptomycin/pharmacology
5.
J Biochem ; 130(6): 727-30, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11726270

ABSTRACT

At the initiation of chromosomal DNA replication, DNA primases synthesize short RNA primers, which are subsequently elongated by DNA polymerases. To understand the structural basis for the primer synthesis by archaeal/eukaryotic-type primases, the gene of the DNA primase from hyperthermophilic archaeon Pyrococcus horikoshii was cloned and overexpressed in Escherichia coli as a fusion protein with a hexa-histidine tag at its amino terminus. The recombinant DNA primase was purified and crystallized by the hanging-drop vapor diffusion method at 293 K, with polyethylene glycol 8000 as the precipitant. The crystals belong to the P3(2)21 space group with unit-cell parameters a = b = 77.8, c = 129.6 A, and alpha = beta = 90 degrees, gamma = 120 degrees. Crystals of the selenomethionine derivative were obtained by means of a cross-seeding method using native crystals. The data for the native and selenomethionine-substituted crystals were collected to 1.8 and 2.2 A resolution, respectively, with synchrotron radiation at SPring-8 under flash-frozen conditions at 100 K. The four wavelength MAD data provided a phase to determine the structure of the primase at 2.2 A resolution.


Subject(s)
Crystallography, X-Ray/methods , DNA Primase/chemistry , Pyrococcus/enzymology , Amino Acid Substitution , Crystallization , DNA Primase/isolation & purification , DNA Primase/metabolism , Protein Conformation , RNA/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Selenomethionine/chemistry
6.
J Mol Biol ; 305(4): 917-26, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11162102

ABSTRACT

The hPar14 protein is a peptidyl prolyl cis/trans isomerase and is a human parvulin homologue. The hPar14 protein shows about 30 % sequence identity with the other human parvulin homologue, hPin1. Here, the solution structure of hPar14 was determined by nuclear magnetic resonance spectroscopy. The N-terminal 35 residues preceding the peptidyl prolyl isomerase domain of hPar14 are unstructured, whereas hPin1 possesses the WW domain at its N terminus. The fold of residues 36-131 of hPar14, which comprises a four-stranded beta-sheet and three alpha-helices, is superimposable onto that of the peptidyl prolyl isomerase domain of hPin1. To investigate the interaction of hPar14 with a substrate, the backbone chemical-shift changes of hPar14 were monitored during titration with a tetra peptide. Met90, Val91, and Phe94 around the N terminus of alpha3 showed large chemical-shift changes. These residues form a hydrophobic patch on the molecular surface of hPar14. Two of these residues are conserved and have been shown to interact with the proline residue of the substrate in hPin1. On the other hand, hPar14 lacks the hPin1 positively charged residues (Lys63, Arg68, and Arg69), which determine the substrate specificity of hPin1 by interacting with phosphorylated Ser or Thr preceding the substrate Pro, and exhibits a different structure in the corresponding region. Therefore, the mechanism determining the substrate specificity seems to be different between hPar14 and hPin1.


Subject(s)
Peptidylprolyl Isomerase/chemistry , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , NIMA-Interacting Peptidylprolyl Isomerase , Nuclear Magnetic Resonance, Biomolecular , Peptidylprolyl Isomerase/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Solutions
8.
Nat Struct Biol ; 7 Suppl: 943-5, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11103994

ABSTRACT

Two major structural genomics projects exist in Japan. The oldest, the RIKEN Structural Genomics Initiative, has two major goals: to determine bacterial, mammalian, and plant protein structures by X-ray crystallography and NMR spectroscopy and to perform functional analyses with the target proteins. The newest, the structural genomics project at the Biological Information Research Center, focuses on human membrane proteins.


Subject(s)
Computational Biology , Genomics , Proteins/chemistry , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell-Free System , Computational Biology/economics , Computational Biology/methods , Crystallography, X-Ray , Genomics/methods , Humans , Internet , Japan , Nuclear Magnetic Resonance, Biomolecular , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Biosynthesis , Protein Conformation , Proteins/genetics , Proteins/metabolism , Structure-Activity Relationship , Workforce
10.
J Biochem ; 127(2): 181-4, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10731682

ABSTRACT

The cell surface antigen CD38 is a multifunctional ectoenzyme that acts as an NAD(+) glycohydrolase, an ADP-ribosyl cyclase, and also a cyclic ADP-ribose hydrolase. The extracellular catalytic domain of CD38 was expressed as a fusion protein with maltose-binding protein, and was crystallized in the complex with a ganglioside, G(T1b), one of the possible physiological inhibitors of this ectoenzyme. Two different crystal forms were obtained using the hanging-drop vapor diffusion method with PEG 10,000 as the precipitant. One form diffracted up to 2.4 A resolution with synchrotron radiation at 100 K, but suffered serious X-ray damage. It belongs to the space group P2(1)2(1)2(1) with unit-cell parameters of a = 47.9, b = 94.9, c = 125.2 A. The other form is a thin plate, but the data sets were successfully collected up to 2.4 A resolution by use of synchrotron radiation at 100 K. The crystals belong to the space group P2(1) with unit-cell parameters of a = 57.4, b = 51.2, c = 101.1 A, and beta = 97.9 degrees, and contain one molecule per asymmetric unit with a VM value of 2.05 A(3)/Da.


Subject(s)
Antigens, CD , Antigens, Differentiation/chemistry , Antigens, Differentiation/metabolism , Gangliosides/chemistry , Gangliosides/metabolism , NAD+ Nucleosidase/chemistry , NAD+ Nucleosidase/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Antigens, Differentiation/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallization , Maltose-Binding Proteins , NAD+ Nucleosidase/genetics , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , X-Ray Diffraction
11.
Oncogene ; 18(13): 2281-90, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10327074

ABSTRACT

To prevent neoplasia, cells of multicellular organisms activate cellular disposal programs such as apoptosis in response to deregulated oncogene expression, making the suppression of such programs an essential step for potentially neoplastic cells to become established as clinically relevant tumors. Since the mutation of ras proto-oncogenes, the most frequently mutated proto-oncogenes in human tumors, is very rare in some tumor types such as glioblastomas and gastric cancers, we hypothesized that mutated ras genes might activate a cell death program that cannot be overcome by these tumor types. Here we show that the expression of oncogenically mutated ras gene induces cellular degeneration accompanied by cytoplasmic vacuoles in human glioma and gastric cancer cell lines. Cells dying as a result of oncogenic Ras expression had relatively well-preserved nuclei that were negative for TUNEL staining. An immunocytochemical analysis demonstrated that the cytoplasmic vacuoles are derived mainly from lysosomes. This oncogenic Ras-induced cell death occurred in the absence of caspase activation, and was not inhibited by the overexpression of anti-apoptotic Bcl-2 protein. These observations suggested that oncogenic Ras-induced cell death is most consistent with a type of programmed cell death designated 'type 2 physiological cell death' or 'autophagic degeneration', and that this cell death is regulated by a molecular mechanism distinct from that of apoptosis. Our findings suggest a possible role for this non-apoptotic cell death in the prevention of neoplasia, and the activation of the non-apoptotic cell death program may become a potential cancer therapy complementing apoptosis-based therapies. In addition, the approach used in this study may be a valuable way to find genetically-regulated cell suicide programs that cannot be overcome by particular tumor types.


Subject(s)
Caspases/physiology , Cell Death/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Genes, ras , Neoplasm Proteins/physiology , Neoplastic Stem Cells/cytology , Proto-Oncogene Proteins p21(ras)/physiology , Brain Neoplasms/pathology , Cell Death/physiology , Cell Nucleus/ultrastructure , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Glioblastoma/pathology , Glioma/pathology , Humans , Lysosomes/ultrastructure , Neoplastic Stem Cells/metabolism , Phagocytosis , Proto-Oncogene Proteins c-bcl-2/physiology , Recombinant Fusion Proteins/physiology , Signal Transduction/genetics , Stomach Neoplasms/pathology , Transfection , Urinary Bladder Neoplasms/pathology , Vacuoles/ultrastructure
12.
Biochemistry ; 38(16): 5103-10, 1999 Apr 20.
Article in English | MEDLINE | ID: mdl-10213614

ABSTRACT

RalGDS is a guanine nucleotide dissociation stimulator for Ral, and one of its homologues is RGL (RalGDS-like). In this study, the effects of mutations of Ras and the Ras-binding domains (RBDs) of RalGDS and RGL on their binding have been systematically examined. The D33A mutation of Ras reduces the abilities to bind RGL-RBD and RalGDS-RBD. To identify the RGL residue interacting with Asp33 of Ras, double-mutant analyses between Ras and RGL-RBD were conducted. For example, the K685A mutation of RGL-RBD has a much smaller effect on the RGL-RBD binding ability of the D33A mutant than on those of other mutants of Ras. Accordingly, it is indicated that the attractive interaction of Asp33 in Ras with Lys685 in RGL-RBD (Lys816 in RalGDS-RBD) contributes to the Ras.RBD association. This interaction is consistent with the crystal structure of the complex of RalGDS-RBD and the E31K Ras mutant [Huang, L., Hofer, F., Martin, G. S., and Kim, S.-H. (1998) Nat. Struct. Biol. 5, 422-426]. This crystal structure exhibits interactions of the mutation-derived Lys31 side chain with three RalGDS residues. Glu31 of Ras discriminates Ras from a Ras-homologue, Rap1, with Lys31, with respect to RalGDS and RGL binding; the E31K mutation of Ras potentiates the abilities to bind RGL-RBD and RalGDS-RBD. To examine the role of Glu31 of the wild-type Ras in the interaction with RGL and RalGDS, double-mutant analyses were conducted. The Ras binding ability of the E689A mutant of RGL-RBD is much stronger than that of the wild-type RGL-RBD, and the E31K mutation of Ras no longer potentiates the Ras binding ability of the E689A mutant. Therefore, the repulsive interaction between Glu31 in Ras and Glu689 in RGL-RBD (Asp820 in RalGDS-RBD) may keep the Ras.RBD association weaker than the Rap1.RBD association, which might be relevant to the regulation of the signaling network.


Subject(s)
GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , ras Proteins/genetics , ras Proteins/metabolism , Alanine/genetics , Amino Acid Sequence , Animals , Aspartic Acid/genetics , Glutamic Acid/genetics , Histidine/genetics , Humans , Lysine/genetics , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Static Electricity , Tyrosine/genetics
13.
J Mol Biol ; 286(1): 219-32, 1999 Feb 12.
Article in English | MEDLINE | ID: mdl-9931261

ABSTRACT

The Ras protein and its homolog, Rap1A, have an identical "effector region" (residues 32-40) preceded by Asp30-Glu31 and Glu30-Lys31, respectively. In the complex of the "Ras-like" E30D/K31E mutant Rap1A with the Ras-binding domain (RBD), residues 51-131 of Raf-1, Glu31 in Rap1A forms a tight salt bridge with Lys84 in Raf-1. However, we have recently found that Raf-1 RBD binding of Ras is indeed reduced by the E31K mutation, but is not affected by the E31A mutation. Here, the "Rap1A-like" D30E/E31K mutant of Ras was prepared and shown to bind the Raf-1 RBD less strongly than wild-type Ras, but slightly more tightly than the E31K mutant. The backbone 1H, 13C, and 15N magnetic resonances of the Raf-1 RBD were assigned in complexes with the wild-type and D30E/E31K mutant Ras proteins in the guanosine 5'-O-(beta,gamma-imidotriphosphate)-bound form. The Lys84 residue in the Raf-1 RBD exhibited a large change in chemical shift upon binding wild-type Ras, suggesting that Lys84 interacts with wild-type Ras. The D30E/E31K mutant of Ras caused nearly the same perturbations in Raf-1 chemical shifts, including that of Lys84. We hypothesized that Glu31 in Ras may not be the major salt bridge partner of Lys84 in Raf-1. A molecular dynamics simulation of a model structure of the Raf-1 RBD.Ras.GTP complex suggested that Lys84 in Raf-1 might instead form a tight salt bridge with Asp33 in Ras. Consistent with this, the D33A mutation in Ras greatly reduced its Raf-I RBD binding activity. We conclude that the major salt bridge partner of Lys84 in Raf-1 may be Asp33 in Ras.


Subject(s)
Proto-Oncogene Proteins c-raf/chemistry , ras Proteins/chemistry , Amino Acid Sequence , Aspartic Acid/metabolism , Computer Simulation , Escherichia coli , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Proto-Oncogene Proteins c-raf/metabolism , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , ras Proteins/genetics , ras Proteins/metabolism
14.
Breast Cancer Res Treat ; 49(1): 51-60, 1998 May.
Article in English | MEDLINE | ID: mdl-9694611

ABSTRACT

Based on estrogen receptor (ER) and menopausal status, operable breast cancer (UICC stage I, II, III-a) patients were randomized for adjuvant endocrine therapy, chemotherapy, and chemoendocrine therapy, and the effects on the relapse-free survival (RFS) and overall survival (OS) were compared. Tamoxifen (TAM) 20 mg/day was administered orally for 2 years after mastectomy as an adjuvant endocrine therapy in postmenopausal patients. In premenopausal patients, oophorectomy (OVEX) was performed before TAM administration. In the chemotherapy arm (CHEM), patients were given 0.06 mg/kg of body weight of mitomycin C (MMC) intravenously, followed by an oral administration of cyclophosphamide (CPA) 100 mg/day in an administration of a 3-month period and a 3-month intermission. This 6-month schedule was repeated 4 times in 2 years. The chemoendocrine arm (CHEM + TAM) was composed of TAM with MMC + CPA chemotherapy. The patients were randomized according to ER and menopausal status. ER-positive patients were randomized to three arms: OVEX +/- TAM, CHEM, and CHEM + TAM. For ER-negative patients there were two arms: CHEM and CHEM + TAM. 1579 patients entered the trial between September 1978 and December 1991, with median follow-up of 8.2 years. In ER-positive, premenopausal patients, there were no significant differences in RFS or OS among OVEX + TAM, MMC + CPA, TAM + MMC + CPA arms. On the contrary, in ER-positive, postmenopausal patients, the chemoendocrine therapy showed a significantly higher RFS (p = 0.0400) and OS (p = 0.0187) as compared with TAM to chemotherapy alone. There were no significant differences in RFS or OS by addition of TAM on the chemotherapy, in both pre- and post-menopausal ER-negative patients. It was concluded that in ER-positive premenopausal breast cancer, endocrine therapy alone may be equivalent in prolonging RFS and OS to chemotherapy or chemoendocrine therapy, and that ER-positive postmenopausal breast cancer may be better controlled with the combination of TAM and chemotherapy, as compared to TAM or chemotherapy alone. The importance of stratification of operable breast cancer by ER and menopausal status, as well as the direct comparisons of different treatments, were stressed.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Receptors, Estrogen/analysis , Tamoxifen/therapeutic use , Adult , Aged , Antineoplastic Agents, Hormonal/administration & dosage , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant , Cyclophosphamide/administration & dosage , Female , Humans , Menopause , Middle Aged , Mitomycin/administration & dosage , Prospective Studies , Tamoxifen/administration & dosage
15.
Oncogene ; 16(20): 2597-607, 1998 May.
Article in English | MEDLINE | ID: mdl-9632136

ABSTRACT

A fragment consisting of residues 584-1071 of the mouse Son-of-sevenless 1 (mSos1) protein was found to be sufficient for stimulation of the guanine nucleotide exchange of Ras in vitro, which defines the CDC25 homology (CDC25H) domain of mSos1. Furthermore, we found that the CDC25H-domain fragment activated the extracellular signal-regulated protein kinases (ERKs), and was mainly membrane localized, when expressed in unstimulated human embryonic kidney 293 cells. Then, we examined the roles of other mSos1 domains in autoinhibition of the CDC25H-domain functions in unstimulated cellular environments. First, longer fragments that have the CDC25H domain and the following proline-rich Grb2-binding domain exhibited negligible membrane localization, and accordingly much lower ERK-activation activities, under serum-starved conditions. On the other hand, the preceding Pleckstrin-homology (PH) domain affects neither the ERK-activation activity nor the membrane-localization activity of the CDC25H domain. By contrast, the cells expressing a fragment containing the Dbl homology (DH) domain in addition to the PH and CDC25H domains exhibited remarkably low ERK activities under serum-starved conditions. This autoinhibitory effect of the DH domain on the CDC25H-domain function was shown to be relieved when cells were stimulated with epidermal growth factor. The DH-domain extension affected neither the in vitro guanine nucleotide exchange activity nor the membrane-localization activity of the CDC25H domain. Therefore, one of the roles of the DH domain is to exert an autoinhibition over the CDC25H-domain function on the cell membrane, in the absence, but not in the presence, of extracellular stimuli.


Subject(s)
Gene Expression Regulation , Genes, ras , Membrane Proteins/chemistry , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins/chemistry , Cells, Cultured , Enzyme Activation , Escherichia coli/genetics , Guanine Nucleotides/metabolism , Humans , Kidney , Mice , Phosphoprotein Phosphatases/chemistry , Plasmids , Recombinant Fusion Proteins , Son of Sevenless Proteins , Transfection , ras-GRF1
16.
J Biol Chem ; 273(13): 7737-42, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9516482

ABSTRACT

The Ras and Rap1A proteins can bind to the Raf and RalGDS families. Ras and Rap1A have Glu and Lys, respectively, at position 31. In the present study, we analyzed the effects of mutating the Glu at position 31 of the c-Ha-Ras protein to Asp, Ala, Arg, and Lys on the interactions with Raf-1 and RalGDS. The Ras-binding domain (RBD) of Raf-1 binds the E31R and E31K Ras mutants less tightly than the wild-type, E31A, and E31D Ras proteins; the introduction of the positively charged Lys or Arg residue at position 31 specifically impairs the binding of Ras with the Raf-1 RBD. On the other hand, the ability of the oncogenic RasG12V protein to activate Raf-1 in HEK293 cells was only partially reduced by the E31R mutation but was drastically impaired by the E31K mutation. Correspondingly, RasG12V(E31K) as well as Rap1A, but not RasG12V(E31R), exhibited abnormally tight binding with the cysteine-rich domain of Raf-1. On the other hand, the E31A, E31R, and E31K mutations, but not the E31D mutation, enhanced the RalGDS RBD-binding activity of Ras, indicating that the negative charge at position 31 of Ras is particularly unfavorable to the interaction with the RalGDS RBD. RasG12V(E31K), RasG12V(E31A), and Rap1A stimulate the RalGDS action more efficiently than the wild-type Ras in the liposome reconstitution assay. All of these results clearly show that the sharp contrast between the characteristics of Ras and Rap1A, with respect to the interactions with Raf-1 and RalGDS, depends on their residues at position 31.


Subject(s)
GTP-Binding Proteins/metabolism , Glutamic Acid/metabolism , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism , 3T3 Cells , Animals , Enzyme Activation , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Mice , Mitogen-Activated Protein Kinase Kinases , PC12 Cells , Point Mutation , Protein Binding , Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/chemistry , Rats , Structure-Activity Relationship , ral Guanine Nucleotide Exchange Factor , rap GTP-Binding Proteins , ras Proteins/chemistry , ras Proteins/genetics
17.
FEBS Lett ; 441(3): 413-8, 1998 Dec 28.
Article in English | MEDLINE | ID: mdl-9891982

ABSTRACT

The RGL protein, a homolog of the Ral GDP dissociation stimulator (RalGDS), has been identified as a downstream effector of Ras. In the present study, the solution structure of the Ras-binding domain of RGL (RGL-RBD) was determined by NMR spectroscopy. The overall fold of RGL-RBD consists of a five-stranded beta-sheet and two alpha-helices, which is the same topology as that of RalGDS-RBD. The backbone chemical shift perturbation of RGL-RBD upon interaction with the GTP analog-bound Ras was also examined. The solution structure of RGL-RBD, especially around some of the Ras-interacting residues, is appreciably different from that of RalGDS-RBD.


Subject(s)
GTP-Binding Proteins/chemistry , Guanine Nucleotide Exchange Factors , ras Proteins/metabolism , Amino Acid Sequence , GTP-Binding Proteins/metabolism , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Solutions
18.
FEBS Lett ; 441(2): 322-6, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9883908

ABSTRACT

RNA aptamers that bind to the Ras-binding domain (RBD) of a proto-oncogene product, Raf-1, were isolated from a pool of random sequences using a glutathione S-transferase-fused RBD (GST-RBD). The RNA molecules bind to the GST-RBD, but not to GST, with dissociation constants of about 300 nM. In contrast, these RNA aptamers do not bind to the Ras-binding domain of the RGL protein, which is also known to be activated by Ras. The aptamers actually compete with Ras for binding to the Raf-1 RBD. The anti-Raf-1 aptamers may be used to specifically inhibit the Ras-Raf interaction in the complicated signaling network in mammalian cells.


Subject(s)
Biopolymers/metabolism , Proto-Oncogene Proteins c-raf/metabolism , RNA/metabolism , ras Proteins/metabolism , Base Sequence , Biopolymers/chemistry , Biopolymers/genetics , Cloning, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Proto-Oncogene Proteins c-raf/chemistry , RNA/chemistry , RNA/genetics , Sequence Homology, Nucleic Acid
19.
Oncogene ; 15(6): 619-27, 1997 Aug 07.
Article in English | MEDLINE | ID: mdl-9264402

ABSTRACT

The Ras signal transduction pathway is activated by a number of hematopoietic cytokines and is implicated in the prevention of apoptotic death in hematopoietic cells. Recent studies have provided evidence that the downstream of Ras is highly divergent and several independent pathways appear to mediate distinct biological functions of Ras. In the present study, we investigated the downstream pathway(s) of Ras responsible for the maintenance of hematopoietic cell survival by using various mutants of signaling molecules. Activation of the Raf/MAPK pathway in interleukin (IL) 3-dependent cells by expression of an oncogenic Raf or a Ras mutant (G12V/T35S) prevented apoptosis following IL-3 deprivation. In contrast, another Ras mutant (G12V/V45E), which is apparently incapable of activating MAPK, efficiently blocked apoptosis as well. It is therefore likely that the activation of the Raf/MAPK pathway is not an absolute requirement for the prevention of apoptosis, and there appears to be a Raf/MAPK-independent pathway that contributes to hematopoietic cell survival. Since Ras(G12V/V45E) was able to cause the phosphorylation of p70/S6 kinase, we inhibited the S6 kinase pathway by rapamycin and by wortmannin, and found that the anti-apoptotic function of Ras(G12V/V45E), but not of Ras(G12V), was critically influenced by both inhibitors. These results indicate that the Raf/MAPK and a rapamycin/wortmannin-sensitive pathways mediate Ras function to prevent apoptotic death in hematopoietic cells.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , MAP Kinase Kinase Kinases , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/physiology , Androstadienes/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Survival , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibroblasts , Interleukin-3/metabolism , Mice , Polyenes/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Recombination, Genetic , Ribosomal Protein S6 Kinases , Signal Transduction , Sirolimus , Transfection , Wortmannin
20.
Biochemistry ; 36(30): 9109-19, 1997 Jul 29.
Article in English | MEDLINE | ID: mdl-9230043

ABSTRACT

The backbone 1H, 13C, and 15N resonances of the c-Ha-Ras protein [a truncated version consisting of residues 1-171, Ras(1-171)] bound with GMPPNP (a slowly hydrolyzable analogue of GTP) were assigned and compared with those of the GDP-bound Ras(1-171). The backbone amide resonances of amino acid residues 10-13, 21, 31-39, 57-64, and 71 of Ras(1-171).GMPPNP, but not those of Ras(1-171).GDP, were extremely broadened, whereas other residues of Ras(1-171).GMPPNP exhibited amide resonances nearly as sharp as those of Ras(1-171). GDP. The residues exhibiting the extreme broadening, except for residues 21 and 71, are localized in three functional loop regions [loops L1, L2 (switch I), and L4 (switch II)], which are involved in hydrolysis of GTP and interactions with other proteins. From the temperature and magnetic field strength dependencies of the backbone amide resonance intensities, the extreme broadening was ascribed to the exchange at an intermediate rate on the NMR time scale. It was shown that the Ras(1-171) protein bound with GTP or GTPgammaS (another slowly hydrolyzable analogue of GTP) exhibits the same type of broadening. Therefore, it is a characteristic feature of the GTP-bound form of Ras that the L1, L2, and L4 loop regions, but not other regions, are in a rather slow interconversion between two or more stable conformers. This phenomenon, termed a "regional polysterism", of these loop regions may be related with their multifunctionality: the GTP-dependent interactions with several downstream target groups such as the Raf and RalGDS families and also with the GTPase activating protein (GAP) family. In fact, the binding of Ras(1-171).GMPPNP with the Ras-binding domain (residues 51-131) of c-Raf-1 was shown to eliminate the regional polysterism nearly completely. It was indicated, therefore, that each target/regulator selects its appropriate conformer among those presented by the "polysteric" binding interface of Ras. As the downstream target groups exhibit no apparent sequence homology to each other, it is possible that one target group prefers a conformer different from that preferred by another group. The involvement of loop L1 in the regional polysterism might suggest that the negative regulators, GAPs, bind to the polysteric binding interface (loops L2 and L4) of Ras and cooperatively select a conformer suitable for transition of the GTPase catalytic center, involving loops L1 and L4, into the highly active state.


Subject(s)
Guanosine Triphosphate/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Carbon Isotopes , Crystallography, X-Ray , Guanosine 5'-O-(3-Thiotriphosphate)/analogs & derivatives , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , Humans , Hydrogen , Magnetic Resonance Spectroscopy , Nitrogen Isotopes , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , Proto-Oncogene Proteins p21(ras)/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...